SciELO - Scientific Electronic Library Online

 
vol.46 issue4Validity of the Pulmonary Embolism Rule-Out Criteria (PERC) for ruling out pulmonary embolism in low-risk patients at high altitudesIt is time to adapt the definition of chronic kidney disease according to age author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

Related links

  • On index processCited by Google
  • Have no similar articlesSimilars in SciELO
  • On index processSimilars in Google

Share


Acta Medica Colombiana

Print version ISSN 0120-2448

Acta Med Colomb vol.46 no.4 Bogotá Oct./Dec. 2021  Epub Apr 28, 2022

https://doi.org/10.36104/amc.2021.2108 

Review

Current treatment for heart failure with reduced ejection fraction. A systematic review of the new therapies

JAIRO ALFONSO GÁNDARA-RICARDOa 

EDISON MUÑOZ-ORTIZb 

OSWALDO ENRIQUE AGUILAR-MOLINAc 

KAREN GARCÍA-RUEDAc 

SANTIAGO GIRALDO-RAMÍREZc 

JHON FREDY SALAMANCA-MONTILLAc 

JUAN MANUEL SÉNIOR-SÁNCHEZd  * 

a Cardiólogo. Sección de Cardiología, Departamento de Medicina Interna Universidad de Antioquia. Clínica de Falla Cardiaca Hospital Universitario San Vicente Fundación. Grupo para el Estudio de las Enfermedades Cardiovasculares Universidad de Antioquia; Medellín, Colombia.

b Cardiólogo. Sección de Cardiología, Departamento de Medicina Interna Universidad de Antioquia. Clínica de Falla Cardiaca Hospital Universitario San Vicente Fundación. Grupo para el Estudio de las Enfermedades Cardiovasculares Universidad de Antioquia; Medellín, Colombia.

c Residentes de Cardiología Clínica, Sección de Cardiología, Departamento de Medicina Interna, Universidad de Antioquia y Pertenecientes al Grupo para el Estudio de las Enfermedades Cardiovasculares Universidad de Antioquia; Medellín, Colombia.

d MSc en Epidemiología Clínica. Coordinador de Posgrado de Cardiología Clínica y Cardiología Intervencionista, Sección de Cardiología, Departamento de Medicina Interna Universidad de Antioquia. Clínica de Falla Cardiaca Hospital Universitario San Vicente Fundación. Grupo para el Estudio de las Enfermedades Cardiovasculares Universidad de Antioquia. Medellín (Colombia).


Abstract

Introduction:

heart failure with reduced ejection fraction has a growing therapeutic arsenal. Thus, the indications for each therapy must be refined.

Methods:

a systematic review was performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, to update the systematic search performed in the development of the "Clinical Practice Guidelines for Prevention, Diagnosis, Treatment and Rehabilitation" (CPG) of the Colombian Ministry of Health.

Results:

six new clinical trials were found which substantially modify the main recommendations of the CPG. Angiotensin receptor antagonists combined with neprilysin inhibitors (ARNI), sodium-glucose cotransporter 2 (SGLT2) inhibitors, betablockers and mineralocorticoid receptor antagonists (MRA) are now the main core of treatment for patients with heart failure with reduced ejection fraction. Other therapeutic options should be considered after beginning and titrating the doses of these four medications.

Discussion:

given the robustness of the evaluating studies, the proposed practical scheme, as the central core with four fundamental therapeutic strategies, will improve the treatment of patients with heart failure and allow the stepwise inclusion of other alternatives, plotted as orbits, to impact on other individual outcomes. (Acta Med Colomb 2021; 46. DOI:https://doi.org/10.36104/amc.2021.2108).

Keywords: heart failure; epidemiology; mortality

Resumen

Introducción:

la falla cardiaca con fracción de eyección reducida dispone de un arsenal terapéutico cada vez más amplio por lo que se requiere refinar las indicaciones de cada una de ellas.

Métodos:

se realizó una revisión sistemática siguiendo las directrices de Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA), para actualizar la búsqueda sistemática realizada para el desarrollo de la "Guía de Practica Clínica para la prevención, diagnóstico, tratamiento y rehabilitación" (GPC) del Ministerio de Salud de Colombia.

Resultados:

se detectaron seis ensayos clínicos nuevos que modifican sustancialmente las recomendaciones principales de la GPC. Los antagonistas del receptor de angiotensina combinado con inhibidor de neprilisina (ARNI), los inhibidores del cotransportador sodio glucosa-2 (ISGLT-2), los betabloqueadores y los antagonistas mineralocorticoides (ARM) se convierten en el núcleo principal del tratamiento de los pacientes con falla cardiaca con fracción de eyección reducida. Otras opciones terapéuticas deben ser consideradas después de iniciar y titular las dosis de estos cuatro medicamentos.

Discusión:

el esquema práctico propuesto, como núcleo central con cuatro estrategias terapéuticas fundamentales, dada la robustez de los resultados de los ensayos en que fueron evaluados, permitirá mejorar el tratamiento de los pacientes con falla cardiaca e incluir en forma escalonada con el uso de otras alternativas, graficado como órbitas, para impactar otros desenlaces individuales. (Acta Med Colomb 2021; 46. DOI:https://doi.org/10.36104/amc.2021.2108).

Palabras clave: falla cardiaca; epidemiología; tratamiento; mortalidad

Introduction

Heart failure (HF) is a clinical syndrome characterized by typical signs and symptoms in addition to the objective evidence of a structural or functional abnormality of the heart 1. It is associated with significant morbidity and mortality, with one-year mortality rates of 7.2% and hospitalization rates of 31.9% in patients with chronic HF, and these outcomes increasing to 17.4% mortality and 43.9% hospitalization in patients with acute HF 2. Five-year survival is 56.7% (95% CI 54-59.4%) and 10-year survival is 34.9% (95% CI 24-46.8%) 3.

Traditionally, left ventricular ejection fraction (LVEF) has been the most commonly used classification method, due to its practical implications for determining an appropriate treatment strategy, although over the last few years the measurement of global longitudinal strain and myocardial contraction fraction have gained ground 4. The European Society of Cardiology classifies it in three categories according to LVEF: HF with preserved LVEF when it is equal to or greater than 50%, HR with mildly reduced LVEF if it is 40-49%, and HF with reduced LVEF (HF-rLVEF) when it is less than 40% 5.

Heart failure with preserved and mid-range LVEF has no specific evidenced-based treatment to date, and its treatment is aimed at managing risk factors and controlling comorbidities 1,2,5, while HF-rLVEF has clearly evidence-based treatments with an increasingly broad therapeutic arsenal, and currently even offers new treatment paradigms 6. Thus we, in this systematic review of the literature, deal with the current treatment of HF-rLVEF.

Data collection

The study design is a systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines 7. The systematic search performed in drafting the "Clinical Practice Guidelines for the Prevention, Diagnosis, Treatment and Rehabilitation of Heart Failure in People Over the Age of 18, Classification B, C and D" 1 was updated.

Objective

The population, intervention, control, and outcome (PICO) questions were:

1. In patients over the age of 18 with HF-rLVEF, does pharmacological or interventional treatment or treatment with devices decrease the combined primary outcome - mortality and hospitalizations for any cause or for cardiovascular causes - compared with a placebo or active treatment or not using them?

Elegibility criteria

Studies were selected by two investigators, and differences were resolved by consensus. The included studies were randomized clinical trials which compared the use of pharmacological or interventional treatment or devices in HF-rLVEF (defined as LVEF less than 40%) with placebo, active treatment or, in the case of devices, with not using them, and which reported relevant combined outcomes: mortality from any cause or cardiovascular mortality, hospitalizations for any cause or for HF. The baseline characteristics and the outcomes evaluated were extracted from each study.

Search strategy

A systematic search was performed in the following bibliographic databases: PubMed, Embase, CENTRAL, DARE, Epistemonikos, SciELO, LILACS and OpenGrey, along with the web pages of clinical trial registries such as Clinical-Trials. The manual search included international cardiology congress pages, web sites and cardiology electronic media like Twitter from institutions such as the European Society of Cardiology, the American Heart Association, the Canadian Cardiovascular Society and the American College of Cardiology, as well as primary studies included in these societies' clinical practice guidelines and those of the National Institute for Health Care Excellence (NICE) and meta-analyses. The update included studies in English from January 2014 to December 2020.

A general and manual search was performed including the following terms: "heart failure" or "congestive heart failure" or "left ventricular dysfunction" or "reduced ejection fraction" or "systolic heart failure" and "treatment" "management" or "drug therapy" and randomized controlled trial or controlled clinical trial or randomized or randomly or trial".

Statistical analysis

In line with the individual characteristics of the included studies, the heterogeneity among them and the fact that they act as complementary therapies, the study group consensus was to perform a systematic review rather than a meta-analysis. The individual papers were analyzed according to their baseline characteristics and defined outcomes, which are presented in tables for comparison. The risk of bias was assessed using the strategy proposed by Cochrane, and the fragility index was calculated for new studies along with a comparison with the classic studies using the outcome of mortality from any cause. The fragility index is defined as the minimum number of events that would have to change to a "non-event" status in order to change from a significant to a nonsignificant result, as evidence of the robustness of the studies, since many of these depend on a difference of three or fewer events; its interpretation shows that the smaller the index, the more fragile the result 8.

Results

The systematic and manual search recovered 1,424 results, of which only original clinical trials were analyzed, excluding publications analyzing individual subgroups or secondary outcomes or subrogates. New studies of angiotensin converting enzyme (ACE) inhibitors or angiotensin II receptor blockers (ARBs) or beta blockers were not recovered, nor were new studies of mineralocorticoid receptor antagonists (MRAs) or ivabradine.

The risk of bias for the six new studies analyzed was considered to be low for all the evaluated aspects: generation of the allocation sequence, allocation concealment, blinding of participants and personnel, blinding of outcomes, incomplete outcomes and selective reporting (Table 1).

Table 1 Risk of bias in the clinical trials. 

Study Random sequence generation Allocation concealment Blinding of participants and personnel Blinding of outcome assessment Incomplete outcome data Selective reporting
PARADIGM HF Low Low Low Low Low Low
DAPA HF Low Low Low Low Low Low
VICTORIA Low Low Low Low Low Low
EMPEROR REDUCED Low Low Low Low Low Low
GALACTIC HF Low Low Low Low Low Low
SOLOIST WHF Low Low Low Low Low Low

The fragility index shows robust results for sacubitril/ valsartan for both the primary combined outcome as well as for cardiovascular death, death from a cardiovascular source and hospitalization for HF. For dapagliflozin, the results are robust for the primary combined outcome and hospitalization for HF. The empagliflozin study showed robust results for the primary combined outcome and hospitalization for HF; for sotagliflozin it is robust for the primary outcome; and vericiguat and omecamtiv mecarbil had very low fragility indices, especially the latter one (Table 2).

Table 2 Fragility index for all the outcomes in the new trials included. 

Fragility index PARADIGM HF Active treatment/ superiority LCZ696/ enalapril DAPA HF Placebo/superiority Dapagliflozin/placebo VICTORIA Placebo/superiority Vericiguat/placebo EMPEROR R Placebo/superiority Empagliflozin/placebo GALACTIC HF Placebo/superiority Omecamtiv/placebo SOLOIST WHF Placebo/superiority Sotagliflozin/placebo
Primary outcome 118 62 8 50 1 72
Cardiovascular death 66 5 0 0 0 0
Total death 49 8 0 0 0 0
Hospitalization for heart failure 54 43 0 50 0 NA

A comparative analysis of the classic and new studies was performed for the outcome of mortality (Table 3). The results showed estimates below 1 in almost all cases, except for valsartan, losartan and omecamtiv mecarbil. Those with a 95% CI above 1 were valsartan and candesartan in the CHARM Added trial, losartan in ELITE II and HEAAL, carvedilol in ANZ, nebivolol, digoxin, ivabradine vericiguat, empagliflozin, omecamtiv mecarbil and sotagliflozin. The fragility indices were robust for beta blockers, spironolactone and sacubitril/valsartan.

Table 3 Comparison of the fragility index of classic HF trials. 

Study Total mortality Estimated 95% CI Fragility index
CONSENSUS 1 year • 36 vs. 52% RR 0.69 (0.52-0.91) 7
SOLVD T 2 years • 35.1 vs. 39.7% RR 0.84 (0.74-0.95) 10
Val-HeFT 2 years • 19.7 vs. 19.4% RR 1.02 (0.88-1.18) 0
CHARM Alternative 3 years • 26.1 vs. 29.1% aHR 0.83 (0.7-0.99) 0
CHARM Added 3.5 years • 29.5 vs. 32.4% HR 0.89 (0.77-1.02) 0
ELITE II 1.5 years • 17.7 vs. 15.9 % HR 1.13 (0.95-1.35) 0
HEAAL 4.7 years • 33 vs. 34.8 % HR 0.94 (0.84-1.04) 0
CIBIS II 1.3 years • 12 vs. 17% HR 0.66 (0.54-0.81) 37
MERIT HF 1 year • 7.2 vs. 11% RR 0.66 (0.53-0.81) 34
COPERNICUS 10.4 m • 11.4 vs. 18.5% RR 0.66 (0.53-0.81) 30
ANZ CARVEDILOL 1.6 years • 9.7 vs. 12.5% RR 0.76 (0.44-1.32) 0
COMET 5 years • 34 vs. 40% HR 0.83 (0.74-0.93) 33
SENIORS 1.75 years • 15.8 vs. 18.1% HR 0.88 (0.71-1.08) 0
RALES 2 years • 35 vs. 46% RR 0.70 (0.6-0.82) 54
EMPHASIS HF 1.75 years • 12.5 vs. 15.5% HR 0.76 (0.62-0.93) 5
DIG 3 years • 34.8 vs. 35.1% RR 0.99 (0.91-1.07) 0
A-HeFT 10 m • 6.2 vs. 10.2% HR 0.57 (no CI) RR 0.63 (0.41-0.96) 3
SHIFT 2 years • 16 vs. 17% HR 0.9 (0.8-1.02) 0
PARADIGM 2.25 years • 17 vs. 19.8% HR 0.84 (0.76-0.93) 49
DAPA HF 18.2 m • 11.6 vs. 13.9% HR 0.83 (0.71 to 0.97) 8
VICTORIA 10.8 m • 20.3 vs. 21.2% HR 0.95 (0.84-1.07) 0
EMPEROR R 16 m • 13.4 vs. 14.2% HR 0.92 (0.77 to 1.10) 0
GALACTIC HF 21.8 m • 25.9 vs. 25.9% HR 1.00 (0.92 to 1.09) 0
SOLOIST WHF 9 m • 13.5 vs. 16.3% HR 0.82 (0.59-1.14) 0

Pharmacological treatment

Angiotensin converting enzyme (ACE) inhibitors

The ACE inhibitors decrease the concentration of angiotensin II and aldosterone, increase renin release and the concentration of angiotensin 9-12, decrease the plasma concentration of epinephrine, norepinephrine and vasopressin, and increase the production of bradykinin 10,13,14.

The CONSENSUS Trial 15 with enalapril was the first drug (1986) to show reduced mortality in HF, in patients in New York Heart Association (NYHA) class IV. Six-month mortality significantly reduced in the group receiving enalapril (44% vs. 26%). Since then, many studies have shown that RAAS inhibitors reduce morbidity and mortality in HF-rLVEF, with reductions in mortality from all causes ranging from 20-30% 16-20.

Angiotensin II receptor blockers (ARBs)

The clinical trial results indicate that ARBs are not superior to ACE inhibitors, but are an alternative for patients with intolerance. Clinical trials have evaluated the efficacy not only of losartan (OPTIME-ELITE II), but also of valsartan (Val-HeFT) and candesartan (CHARM) in patients with HF, in terms of cardiovascular morbidity and mortality 21-24. However, they have not been able to surpass the ACE inhibitors in consistently reducing morbidity and mortality; their main advantage lies in a better tolerance. Their use as an alternative treatment is based on the results of the CHARM-Alternative study in patients with symptomatic HF and LVEF <40% who were not taking ACE inhibitors due to intolerance. In these patients, the use of candesartan achieved a significant reduction in both mortality and the number of hospitalizations for HF 25.

Beta blockers

Beta blockers should be part of the treatment of all chronic, stable patients, and they should be started early after compensating patients with acute, newly diagnosed HF 1,5. They have proven benefits, regardless of the etiology of the HF. These include symptom relief and improved quality of life, reduced risk of hospitalization and incidence of sudden death, as well as improved survival. In addition, they have significant effects on reverse ventricular remodeling, with LVEF recovery 26-30. Beta blocker treatment should be used with caution in patients with borderline BP, recent inotrope treatment or NYHA functional class IV.

A recent meta-analysis raises questions about the effect of beta blockers on patients with HF and atrial fibrillation (AF), as they have a different impact in these patients than in patients with a sinus rhythm 31. The recent RATE AF clinical trial shows that digoxin, compared with bisoprolol, has similar effectivity, but with fewer adverse events 32.

Mineralocorticoid receptor antagonists (MRAs)

The MRAs (spironolactone and eplerenone) reduce the aldosterone escape phenomenon, contribute to the RAAS block 33-35 and reduce mortality by 15 to 30%. Furthermore, they decreased hospitalizations for HF by 15 to 40% in three randomized clinical trials (RALES, EMPHASIS, EPHESUS) 36-38, in patients with chronic HF-rLVEF, including patients who had had a myocardial infarction, excluding patients with a baseline serum creatinine greater than 2.5 mg/dL (or an estimated glomerular filtration rate [GFR] <30 mL/min/1.73 m2) or a serum potassium level >5.0 mEq/L.

Recent evidence, using indirect comparison, suggests greater survival benefits with a starting strategy using the three agents compared with conventional therapy 39. New drugs such as finerenone have shown benefits in a Phase 2 trial (ARTS-HF) and in type 2 diabetic patients with chronic kidney disease 40-41.

Angiotensin receptor-neprilysin inhibitors (ARNIs)

Initially, as a complement to the neurohormonal block, an attempt was made to raise natriuretic peptide levels with nesiritide and carperitide, with discouraging results 42-45. Subsequently, isolated neprilysin inhibition was attempted with racecadotril, candoxatrilat and ecadotril, which was unsuccessful, as their effect was rapidly lost 46. The next step was to use a new group known as vasopeptidase inhibitors which have a dual mechanism, inhibiting neutral endopeptidases (such as neprilysin) and ACE (omapatrilat). The OVERTURE trial showed noninferiority compared with enalapril, but a greater number of patients with angioedema [24 (0.8%) vs. 14 (0.5%)] 47, due to neprilysin degrading aminopeptidase P which, like ACE, degrades bradykinin.

Finally, sacubitril (a neprilysin inhibitor) was combined with an ARB-II (valsartan), which was tested in the PARADIGM HF trial 48 in patients with LVEF <35%, with proven tolerance to ACE inhibitors or ARBs, NYHA ≥II, elevated natriuretic peptides, systolic arterial pressure (SAP) > 100 mmHg, GFR > 30 mL/min/1.73m2 and potassium ≤5.2 mEq/L. Sacubitril/valsartan showed superiority in doses of 200 mg every 12 hours, compared with enalapril at 10 mg every 12 hours, in cardiovascular mortality, including sudden death and worsening HF (HR 0.8, CI 0.73-0.87) and hospitalizations for HF (HR 0.79, CI 0.71-0.89).

The PIONEER HF trial 49 proved the safety of beginning this drug in uncompensated HF in patients with LVEF <40%, BNP ≥400 ng/L or NT-proBNP ≥1,600 ng/L, no inotrope requirement in the last 24 hours, nor use of vasodilators, SAP <100 mmHg or increased diuretics in the last six hours. Sacubitril/valsartan showed superiority at a dose of 200 mg every 12 hours compared with enalapril at doses of 10 mg every 12 hours, with regard to decreased NT-ProBNP levels and a reduction in 30-day readmissions as a secondary outcome.

Subsequently, the TITRATION trial was performed to determine the safety of conservative (standard) titration or condensed (aggressive) titration of the dose, with good results in both arms 50. Similarly, the TRANSITION trial 51 showed that initiating sacubitril/valsartan before or after discharge did not affect the achievement of the target dose. Finally, the PROVE HF study 52 found a weak, but statistically significant, relationship between NT-ProBNP levels and the change in LVEF, since the bioavailability of valsartan in the ARNI is 50% greater than that of valsartan alone, which means that 400 mg of the combination contains ~203 mg of valsartan, equivalent to 320 of valsartan sold alone; in addition, it has proven cost-effectiveness (Tables 4-6) 19.

Table 4 Comparison of the new HF treatments. Baseline characteristics. 

Variable PARADIGM HF Active treatment/superiority LCZ696/enalapril DAPA HF Placebo/superiority DAPA/placebo VICTORIA Placebo/superiority Vericiguat/placebo EMPEROR R Placebo/superiority Empagliflozin/placebo GALACTIC HF Placebo/superiority Omecamtiv/placebo SOLOIST WHF Placebo/superiority Sotagliflozin/placebo
Group ARNI ISGLT2 SGCE ISGLT2 CMA ISGLT2
Year 2014 2019 2020 2020 2020 2020
Median follow up 27 months 18 months 11 months 16 months 21.8 months 9 months
Sample size 4,187/4,212 2,373/2,371 2,526/2,524 1,863/1,867 4,120/4,112 608/614
Design RCT RCT RCT RCT RCT RCT
Age (years) 63.8 ± 11.5/63.8±11.3 66.2±11/66.5±10.8 67.5±12.2/67.2±12.2 67.2±10.8/66.5±11.2 64.5±11.3/64.5±11.4 69 (63-76)/70 (64-76)
Average GFR 68±20/68±20 66±19.6/65.5±19.3 61.3±27/61.7±27.3 61.8±21.7/62.2±21.5 58.7 (43.8-73.7)/58.8 (44.3-74.3) 49.2 (39.5-61.2)/50.5 (40.5-64.6)
Ejection fraction (%) 29.6±6.1/29.4±6.3 31.2±6.7/30.9±6.9 29±8.3/28.8±8.3 27.7±6.0/27.2±6.1 26.6±6.3/26.5±6.3 35 (28-47)/35 (28-45)
Ischemic (%) 59.9/60.1 55.5/57.3 59.8/56.8 52.8/50.7 53.2/54
NYHA I/II/III/ 4.3/71.6/23.1/0.8; 5/69.3/24.9/0.6 0/67.7/31.5/0.8; 0/67.4/31.7/1.0 0/58.6/40/1.4; 0.1/59.3/39.4/1.2 0/75.1/24.4/0.5; 0/75/24.4/0.6 0/53.3/43.7/3; 0/52.8/44.1/3 Hospitalized + IV diuretic
Hypertension (%) 70.9/70.5 79.3/79 72.4/72.3
Diabetes (%) 34.7/34.6 41.8/41.8 48.6/45.3 49.8/49.8 40.1/40.3 Type 2 100%
Atrial fibrillation 36.2/37.4 38.6/38 43.5/46.4 35.6/37.8 27.8/26.7
Beta blocker (%) 93.1/92.9 96/96.2 93.2/93 94.7/94.7 94.2/94.4 92.8/91.4
Aldosterone antagonist (%) 54.2/57 71.5/70.6 69.3/71.4 70.1/72.6 77.6/77.8 66.3/62.7
ACE inhibitor/ ARB (%) 84.5/82.8 73.3/73.6 70.5/68.9 87/87 82/83
Sacubitril/valsartan (%) 10.5/10.9 14.3/14.7 18.3/20.7 19.9/19 15.3/18.2
ICD 14.9/14.7 26.2/26.1 27.6/27.9 31.0/31.8 32.2/31.3
CRT 7/6.7 8/6.9 14.7/14.6 11.8/11.9 14.4/13.8
NT-proBNP pg/mL 1,631/1,594 1,428/1,446 2,803.5/2,821 1,887/1,926 1,977/2,025 1,816/1,741

Table 5 Comparison of the new HF treatments. Outcomes. 

Outcome PARADIGM HF Active treatment/superiority LCZ696/enalapril DAPA HF Placebo/superiority DAPA/placebo VICTORIA Placebo/superiority Vericiguat/placebo EMPEROR R Placebo/superiority Empagliflozin/placebo GALACTIC HF Placebo/superiority Omecamtiv/placebo SOLOIST WHF Placebo/superiority Sotagliflozin/placebo
Primary combined (%, HR, events per 100 patient years) 21.8/26.5 HR 0.80 (0.73-0.87) 16.3/21.2 HR 0.74 (0.65-0.85) 11.6/15.6 35.5/38.5 HR 0.90 (0.82-0.98) 33.6/37.8 19.4/24.7 HR 0.75 ( 0.65-0.86) 15.8/21.0 37/39.1 HR 0.92 (0.86-0.99) 24.2/26.3 40.3/ 57.8 HR 0.67 (0.52-0.85) 51/76.3
CV death 13.3/16.5 HR 0.80 (0.71-0.89) 9.6/11.5 HR 0.82 (0.69-0.98) 6.5/11.5 16.4/17.5 HR 0.93 (0.81-1.06) 12.9/13.9 10/10.8 HR 0.92 (0.75-1.12) 7.6/8.1 19.1/19.4 HR 1.01 (0.92-1.11) 10.9/10.8 8.4/ 9.5 HR 0-84 (0.58-1.22) 10.6/12.5
First hospitalization for heart failure 12.8/15.6 HR 0.79 (0.71-0.89) 9.7/13.4 HR 0.70 (0.59-0.83) 6.9/9.8 27.4/29.6 HR 0.90 (0.81-1.0) 25.9/29.1 13.2/18.3 HR 0.69 (0.59-0.81) 10.7/15.5 27.7/28.7 HR 0.95 (0.87-1.03) 18/19.1
Total hospitalization 39.7/43.5 HR 0.88 (0.82-0.94) HR 0.85 (0.75-0.95) 28.6/30.1 HR 0.93 (0.86-1) 18.7/20.3
Total death 17/19.8 HR 0.84 (0.76-0.93) 11.6/13.9 HR 0.83 (0.71-0.97) 7.9/9.5 20.3/21.2 HR 0.95 (0.84-1.07) 16/16.9 13.4/14.2 HR 0.92 (0.77-1.10) 10.1/ 10.7 25.9/25.9 HR 1 (0.92-1.09) 14.4/14.4 10.7/ 12.4 HR 0.82 (0.59-1.14) 13.5/16.3
KCCQ change "-2.9±0.36/-4.63±0.36" HR 1.64 (0.63-2.65) 6.1±18.6/3.3±19.2 HR 1.18 (1.11-1.26) 5.8±0.4/4.1±0.4 HR 1.7 (0.5-3.0) 5.8±0.3/6.3±0.3 -0.5 (-1.4 to 0.5) 17.7/13.6
Kidney worsening 2.2/2.6 HR 0.86 (0.65-1.13) 1.2/1.6 HR 0.71 (0.44-1.16) 0.8/1.2 1.6/3.1 HR 0.50 (0.32-0.77)

Table 6 Comparison of the new HF treatments. Adverse events. 

Adverse event PARADIGM HF Active treatment/superiority LCZ696/enalapril DAPA HF Placebo/superiority DAPA/placebo VICTORIA Placebo/superiority Vericiguat/placebo EMPEROR R Placebo/superiority Empagliflozin/placebo GALACTIC HF Placebo/superiority Omecamtiv/placebo SOLOIST WHF Placebo/superiority Sotagliflozin/placebo
Symptomatic hypotension (%, p) 14/9.2 p<0.001 0.3/0.5 9.1/7.9 p=0.12 6/4.9
Hypoglycemia 1.5/0.3
Renal adverse event 3.3/4.5 p<0.007 6.5/7.2 p=0.36 2.1/2.2
Hypercalemia 16.1/17.3 p=0.15 0.1/0.2 4.4/5.6
Cough 11.3/14.3 p<0.001 4.4/4.2
Angioedema 0.4/0.2
Volume depletion 7.5/6.8 P=0.4
Syncope 4/3.5 p=0.30
Urinary tract infection 4.8/5.1
Diarrhea 6.1/3.4
Anemia 7.6/5.7
Ischemic cardiac event 4.9/4.6
Severe ventricular arrhythmia 2.9/3.1

Sodium-glucose cotransporter-2 (SGLT-2) inhibitors

Beginning in 2016, with the publication of the EMPAREG-OUTCOME 53 study which found a reduction in hospitalization for HF in patients with diabetes mellitus (DM) receiving empagliflozin treatment, there has been a growing interest in the effect of this group of medications on HF treatment. The proposed mechanism of action of SGLT-2 inhibitors is, first of all, natriuresis and glucosuria which, in addition to decreasing preload and postload, decreases interstitial edema which is related to ventricular hypertrophy, although this effect is neutralized around week 12 of treatment 54. The second proposed effect is decreased BP, secondary to decreased sympathetic tone through direct inhibition of noradrenaline synthesis by blocking renal tyrosine hydroxylase 55. The third mechanism is decreased cardiomyocyte glucose consumption in favor of the use of fatty acids and ketones which, besides being more energy efficient, have the ability to decrease oxidative stress 56. Finally, although there are several ideas to explain improved contractility through calcium metabolism, direct blocking of the NHE1 receptor by SGLT-2 inhibitors and the subsequent reduction in intracellular sodium and calcium, as well as increased mitochondrial calcium, has the greatest evidence 56.

Their clinical effect has been proven in recent years in patients without DM; thus, the DEFINE HF 57 study found that patients with LVEF <40%, NYHA II-III, elevated natriuretic peptides and GFR ≥ 30mL/min/1.73m2 dapagliflozin at 10 mg/day was superior to placebo in decreasing the dual co-primary outcome which included reducing NT-proBNP levels by at least 20% and increasing the Kansas scale by ≥ 5 points (OR 1.8 CI 1.03-3.06), although there were no differences in the other co-primary outcome of the average NT-proBNP levels at 6-12 weeks.

Later, the DAPA-HF 58 trial showed that in patients with LVEF <40% and NYHA ≥II with elevated natriuretic peptides, despite being on optimal therapy, with GFR ≥ 30 mL/min/1.73m2, SAP > 95 mmHg, and without type 1 DM, dapagliflozin was superior at 10 mg/day compared with placebo in the combined outcome of worsening HF (unplanned hospitalization or an urgent visit to the emergency room for intravenous therapy) or cardiovascular death (HR 0.74 CI 0.65-0.85).

The EMPEROR REDUCED 59 study randomized patients with LVEF <30%, NYHA ≥II, and elevated natriuretic peptides (in cases with LVEF between 30 and 40%, hospitalizations for HF in the previous year or very high peptide levels were also required), despite being on optimal therapy, with a GFR ≥20 mL/min/1.73m2 and SAP >100 mmHg. In this study, empagliflozin proved to be superior at 10 mg/day compared with placebo in decreasing hospitalizations for HF or cardiovascular death (HR 0.75 95% CI 0.65-0.86).

Finally, the SOLOIST WHF study in type 2 diabetic patients with decompensated HF randomly assigned stabilized patients, before or within three days of discharge, to 200 mg of sotaglifozin (increasing to 400 mg, depending on side effects) versus placebo. After a median nine-month follow up, it significantly decreased the combined primary outcome (HR 0.67 95% CI 0.52-0.85). This study offers an interesting perspective by suggesting that early initiation of treatment has important effects and, furthermore, the effect is maintained in patients with an LVEF ≥50%, which suggests a promising effect for patients with HF with a slightly reduced and preserved fraction 60, as suggested by the results of the recent EMPEROR PRESERVED (N Engl J Med 2021 DOI: 10.1056/NEJMoa2107038/10.1056) study.

Although, given the consistency of the described evidence, it could be deduced that this is a group effect, the results of the VERTIS CV 61 trial raised doubts in this regard, since the use of ertugliflozin in patients with type 2 diabetes was unable to show superiority to the placebo, but did show non-inferiority, although the population of HF patients included did not exceed 25%. A subsequent report with the prespecified analysis of HF-related events suggested a reduction in hospitalization for HF (RR 0.70 95% CI 0.56-0.87) and the combined outcome of hospitalization for HF/cardiovascular death (RR 0.83 95% CI 0.72-0.96) 62.

Ivabradine

Ivabradine inhibits the sinus node's pacemaker activity by selectively blocking the hyperpolarization channels known as the "funny" (If) channels, decreasing the heart rate of patients with a sinus rhythm without affecting arterial pressure, cardiac contractility or intracardiac conduction 30. Among the studies found on this treatment is the initial background in the BEAUTIFUL 63 study on patients with coronary disease and ventricular dysfunction (LVEF <40%), which found no difference between ivabradine and placebo in the primary outcome (HR 1 95% CI 0.91-1.1) nor in the secondary outcomes which made up the combined primary outcome. However, in the subgroup analysis, in patients with a baseline heart rate ≥70 bpm, it reduced the incidence of hospitalization for myocardial infarction and coronary revascularization, although it did not decrease the primary objective nor overall mortality 64.

The SHIFT 65 study was conducted in patients with chronic HF and LVEF ≤35%, sinus rhythm and a resting heart rate ≥70 bpm. Ivabradine had an impact on the primary outcome of cardiovascular death or hospitalization for HF (HR 0.82 95% CI 0.75-0.90), as well as the outcomes related to HF (hospitalization for HF HR 0.74 95% CI 0.66-0.83; and death from HF HR 0.74 95% CI 0.58-0.94).

Patients treated with ivabradine have an average heart rate reduction of 8 bpm, while a meta-analysis of patients with HF-rLVEF with beta blockers showed a reduction of 12 bpm 66. Cullington et al. 67 showed that the proportion of patients for whom the addition of ivabradine would be indicated according to the SHIFT trial criteria dropped from 19.4% of the total at the initial visit to only 9% at the 12-month follow up, after adequately adjusting the beta blocker dose.

Hydralazine and isosorbide dinitrate

The evidence for the clinical usefulness of hydralazine and isosorbide dinitrate comes initially from the V-HeFT I trial, which compared three treatments: placebo, prazosin, or isosorbide dinitrate and hydralazine. After two years, there was a 34% reduction in the primary outcome of cumulative mortality in the hydralazine and isosorbide dinitrate group compared with placebo (p=0.028, 95% CI 4-54%) 68. Subsequently, the V-HeFT II study compared hydralazine combined with isosorbide dinitrate to enalapril; at two years, there was a significantly higher reduction in mortality from all causes in the enalapril group (ARR 6%, p= 0.016) and the differences in mortality between the two groups were not sustained at 2.5 years (ARR 5.4%, p=0.08), improved peak oxygen consumption and LVEF in the hydralazine-isosorbide dinitrate group compared with enalapril early on, as part of the secondary results 69.

The AHeFT study was carried out based on observations which suggested a greater benefit in African Americans. This study randomized 1,050 black patients with HF in functional class III/IV and ventricular dilation to receive a fixed dose of hydralazine and isosorbide dinitrate (n=518) compared to placebo (n=532) in addition to standard medical care. The study was ended early when it showed that hydralazine combined with isosorbide dinitrate resulted in significantly lower mortality compared with placebo (6.2% vs. 10.2%, p= 0.02). The outcome of side effects was also better in the intervention group, with a 43% reduction in death from any cause (HR 0.57; p=0.01), 33% reduction in the rate of first hospitalizations for HF (16.4% vs. 22.4%, p=0.01), and improved quality of life 70.

Vericiguat

Vericiguat is an oral, soluble guanylate cyclase stimulator which increases cyclic GMP activity, a second messenger involved in regulating cardio-renal and metabolic protective mechanisms. The SOCRATES-REDUCED study randomized 456 patients with HF with LVEF <45% and one recent decompensation episode (<4 weeks). A total of 377 patients finished treatment and there was no difference between the baseline and 12-week NT-proBNP levels between the group treated with vericiguat and the placebo group, with adequate tolerance and no differences in adverse events, with a linear relationship between the dose of vericiguat used and the degree of NT-proBNP reduction 71.

The VICTORIA study involved 5,050 patients with high-risk HF-rLVEF (40% in NYHA functional class III), severe impairment of systolic ventricular function (mean LVEF of 29%), with elevated natriuretic peptides and recent decompensation (80% within the last three months) to receive oral vericiguat with a target dose of 10 mg per day, compared with placebo plus guideline-directed treatment. The study showed a modest reduction in the primary outcome of cardiovascular death or first hospitalization for HF (35.5% vs. 38.5%, HR 0.9 [95% CI, 0.82-0.98, p=0.02]), derived mainly from the effect on hospitalizations for HF or any cause, with no impact on cardiovascular deaths or total deaths. Mean follow up was 10.8 months. Due to its vasodilating properties, there was symptomatic hypotension in 9.1% of the patients compared with 7.9% in the placebo group (p=0.12) and syncope in 4% compared with 3.5% in the placebo group (p=0.30) 72.

Omecamtiv mecarbil

Omecamtiv mecarbil is a selective cardiac myosin activator which increases contractility by binding to an allosteric site, for a faster transition from weak to strong binding, increasing the number of myosin heads bound to an actin filament, generating more strength. In addition, stabilization of the baseline status reduces ATP turnover in the absence of an interaction with actin, which improves energy efficiency.

The Phase 3 GALACTIC HF trial in 8,256 patients with chronic HF with LVEF ≤35% compared omecamtiv mecarbil with placebo and showed a reduction in the primary outcome of HF-related events or cardiovascular death (HR 0.92 95% CI 0.86-0.99), with no impact on cardiovascular mortality or quality of life assessed with the Kansas questionnaire. Despite its modest effect in the total group, the finding of the differential impact according to LVEF, with a median cut-off ≤ 28% generates an interesting hypothesis which should be tested in a subsequent clinical trial 73.

Digoxin

Digoxin, a sodium-potassium ATPase pump inhibitor 74 was evaluated in the DIG study in 6,800 patients with HF and LVEF <45%. The study showed no differences in mortality [34.8% vs. 35.1% (RR 0.99 95% CI 0.91-1.07, p=0.08)], but there was a reduction in total hospitalization and hospitalization for decompensated HF [26.8% vs. 34.7% (RR 0.72 95% CI 0.66-0.79, p=0.001)]. An analysis of secondary outcomes showed that digoxin had the tendency to reduce deaths attributable to worsening HF [RR 0.88 (95% CI, 0.77-1.01, p=0.06)] 75. The prespecified subgroup analysis of high-risk patients (NYHA III-IV, LVEF < 25%, cardiothoracic ratio > 55%) after two years of follow up showed a reduction in the outcome of hospitalization and death from any cause 76.

A Cochrane review showed that digoxin does not reduce mortality from all causes nor from HF, but it does reduce HF symptoms and readmissions by 32% [OR 0.68 (95% CI 0.61-0.75, p < 0.00001)]. The benefits were greater in patients with greater LVEF impairment (<25%) or NYHA functional class III/IV. A subgroup analysis by serum digoxin concentrations showed that patients with levels between 0.5 and 0.8 ng/mL had a 20% reduction in deaths from all causes (HR 0.8 [95% CI 0.68-0.94, p= 0.005]). There were more arrhythmia complications in patients with serum digoxin levels >1.2 ng/mL, especially in patients with kidney disease, who require close monitoring 77.

Diuretics

Their use is mainly based on small, controlled clinical trials in which they have proven to be well tolerated and to alleviate congestion, improving edemas and reducing body weight 78. Likewise, changes in neurohormonal parameters associated with congestion have been shown 63. Loop diuretics are preferred (furosemide, bumetanide, torsemide), although thiazides may be added in diuretic-resistant patients, as well as potassium-sparing diuretics 3,25. The main adverse events associated with their use are fluid and electrolyte disorders and renal dysfunction 79; the long-term effect on outcomes is unclear 80.

Studies of other treatment options for managing congestion, such as arginine vasopressin antagonists, have shown no impact on clinical outcomes, although they do increase urine volume and decrease weight 81,82.

Treatment of comorbidities and devices

Other types of therapies exist, including iron supplementation, management of atrial fibrillation, implantable automatic defibrillators, cardiac resynchronization, cardiac contractility modulation, mitral clips, CardioMEMS and ventricular assist devices which have proven beneficial in specific groups of patients, after optimizing what is considered to be standard treatment 83-129 (Annex 1).

Discussion

Given the current evidence, the papers published especially during the last year, the GPC recommended heart failure algorithm should be updated to introduce new therapies, considering the magnitude of the treatment benefit and that the effect could be considered to be separate from each of the medication groups, supported by the studies' subgroup analyses.

The consistency of studies with ARNI place it as the treatment of choice, replacing ACE inhibitors/ARBs, and reserving these for a few cases, specifically patients who do not tolerate ARNI treatment due to hypotension. Likewise, the SGLT-2 inhibitors, together with the already positioned beta blockers and MRAs complement this first-line therapy. The results with other new medications such as vericiguat and omecamtiv mecarbil are modest and they should be reserved for only a few cases which are not stabilized using standard treatment. The risk of bias was considered to be low in all the items.

The fragility index analysis comparing the classical strategies with the new ones showed robust results for beta blockers, spironolactone and sacubitril/valsartan, and weak results for ACE inhibitors, ARBs, vericiguat and omecamtiv mecarbil, although it should be noted that this may be affected by factors such as the expected effect size, the number of events and the study's power 130.

The proposal was designed as a decision circle with a central nucleus including what is considered to be the pillar of HF treatment (Figure 1), which is enlarged according to certain variables which may be considered or introduced into treatment, according to the clinical progress.

Figure 1 Decision circle for managing HF-rLVEF. 

The central nucleus is organized hierarchically to highlight the fundamental pillars of treatment and provide a general recommendation for initiating and titrating doses according to monitoring and the patient's clinical progress, supported by the mentioned studies. The next orbit contains the interventions to be used after reaching the goals of the basic nucleus which contains four strategies, whose evidential strength is differentiated based on the support of designed clinical trials, the affected outcome and the variables to be considered for its use.

The suggestion is to begin in step one with ARNI (Figure 2); the only available one to date is the sacubitril/valsartan combination. In step 2, SGLT-2 inhibitors plus the beta blocker (Figure 3), and, finally, the MRA (Figure 4).

Figure 2 HF treatment. Step 1. 

Figure 3 HF treatment. Step 2. 

Figure 4 Heart failure treatment. Step 3 

It is important to highlight that the algorithm is conceived in order to begin these treatments, considered to be pillars, early; and that the suggested titration may be modified according to the patient's clinical condition, although it is expected that the use of these recommendations will improve tolerance and adherence, in general, and that low doses of all the groups may be prescribed, to be subsequently increased according to the set sequence of steps 131.

Other therapies, such as cardiac rehabilitation, including an exercise program, vaccination and palliative care should be used concomitantly, ideally within a structured Heart Failure Clinic program. Patients who remain symptomatic despite the use of this strategy should be referred to a heart transplant group for assessment, especially if they are between 18 and 70 years old; have an NYHA functional status III or IV; a peak oxygen consumption (VO2 max) ≤ 14 mL/kg/minute or, with beta blockers, ≤ 12 mL/kg/minute, in patients < 50 years old add VO2 max ≤50% of the expected (predicted) value and in a submaximal test (RER <1.05) use the slope of the ventilatory equivalent of CO2 > 35 on the cardiopulmonary stress test; and are classified as high risk according to the Heart Failure Survival Score or have <80% expected one-year survival on the Seattle Heart Failure Model scale 132.

References

1. Senior JM, Lugo LH, Díaz JS, Muñoz E TN et al. C. Ministerio de Salud y Protección Social. Guía de Práctica Clínica para la prevención, diagnóstico, tratamiento y rehabilitación de la falla cardíaca en población mayor de 18 años clasificación B, C y D. Gpc.minsalud.gov.co, Guía Complet No 53 [GPC en Internet] Edición Bogotá DC El Minist 2016 [consultada 2016]. 2016: Disponible en gpc.minsalud.gov.co. [ Links ]

2. Maggioni AP, Dahlstrom U, Filippatos G, et al. EUR Observational Research Programme: regional differences and 1-year follow-up results of the Heart Failure Pilot Survey (ESC-HF Pilot). Eur J Heart Fail. 2013;15(7):808-817. doi:10.1093/eurjhf/hft050 [ Links ]

3. Jones NR, Roalfe AK, Adoki I, Hobbs FDR, Taylor CJ. Survival of patients with chronic heart failure in the community: a systematic review and meta D analysis. Eur J Heart Fail. 2019;21(11):1306-1325. doi:10.1002/ejhf.1594 [ Links ]

4. Cikes M, Solomon SD. Beyond ejection fraction: an integrative approach for assessment of cardiac structure and function in heart failure. Eur Heart J. 2016;37(21):1642-1650. doi:10.1093/eurheartj/ehv510 [ Links ]

5. McDonagh T, Metra M, Adamo M, Gardner R, Baumbach A et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart Failure. Eur Heart J 2021;42(36):3599-3726. doi: 10.1093/eurheartj/ehab368. [ Links ]

6. Burnett H, Earley A, Voors AA, et al. Thirty Years of Evidence on the Efficacy of Drug Treatments for Chronic Heart Failure With Reduced Ejection Fraction. Circ Hear Fail. 2017;10(1). doi:10.1161/CIRCHEARTFAILURE.116.003529 [ Links ]

7. Liberati A. The PRISMA Statement for Reporting Systematic Reviews and Meta-Analyses of Studies That Evaluate Health Care Interventions: Explanation and Elaboration. Ann Intern Med. 2009;151(4):W. doi:10.7326/0003-4819-151-4-200908180-00136 [ Links ]

8. Walsh M, Srinathan SK, McAuley DF, et al. The statistical significance of randomized controlled trial results is frequently fragile: a case for a Fragility Index. J Clin Epidemiol. 2014;67(6):622-628. doi:10.1016/j.jclinepi.2013.10.019 [ Links ]

9. Ondetti, M. A., Williams, N. J., Sabo, E., Pluscec, J., Weaver, E. R., & Kocy, O. Angiotensin-converting enzyme inhibitors from the venom of Bothrops jararaca. Isolation, elucidation of structure, and synthesis. Biochemistry, 10(22), 4033-4039. [ Links ]

10. Hoyer J, Schulte K-L, Lenz T. Clinical Pharmacokinetics of Angiotensin Converting Enzyme (ACE) Inhibitors in Renal Failure. Clin Pharmacokinet. 1993;24(3):230-254. doi:10.2165/00003088-199324030-00005 [ Links ]

11. Kostis JB. Angiotensin-Converting Enzyme Inhibitors. Am J Hypertens. 1989;2(1):57-64. doi:10.1093/ajh/2.1.57 [ Links ]

12. Francis GS, Benedict C, Johnstone DE, et al. Comparison of neuroendocrine activation in patients with left ventricular dysfunction with and without congestive heart failure. A substudy of the Studies of Left Ventricular Dysfunction (SOLVD). Circulation. 1990;82(5):1724-1729. doi:10.1161/01.CIR.82.5.1724 [ Links ]

13. Swedberg K. Importance of neuroendocrine activation in chronic heart failure. Impact on treatment strategies. Eur J Heart Fail. 2000;2(3):229-233. doi:10.1016/S1388-9842(00)00102-1 [ Links ]

14. Oates JA, Wood AJJ, Williams GH. Converting-Enzyme Inhibitors in the Treatment of Hypertension. N Engl J Med. 1988;319(23):1517-1525. doi:10.1056/NEJM198812083192305 [ Links ]

15. Effects of Enalapril on Mortality in Severe Congestive Heart Failure. N Engl J Med. 1987;316(23):1429-1435. doi:10.1056/NEJM198706043162301 [ Links ]

16. Effect of Enalapril on Survival in Patients with Reduced Left Ventricular Ejection Fractions and Congestive Heart Failure. N Engl J Med. 1991;325(5):293-302. doi:10.1056/NEJM199108013250501 [ Links ]

17. Pfeffer MA, Braunwald E, Moyé LA, et al. Effect of Captopril on Mortality and Morbidity in Patients with Left Ventricular Dysfunction after Myocardial Infarction. N Engl J Med. 1992;327(10):669-677. doi:10.1056/NEJM199209033271001 [ Links ]

18. The Acute Infarction Ramipril Efficacy (AIRE) Study Investigators. Effect of ramipril on mortality and morbidity of survivors of acute myocardial infarction with clinical evidence of heart failure. Lancet. 1993;342(8875). doi:10.1016/0140-6736(93)92693-N [ Links ]

19. Gaziano TA, Fonarow GC, Velazquez EJ, Morrow DA, Braunwald E, Solomon SD. Cost-effectiveness of Sacubitril-Valsartan in Hospitalized Patients Who Have Heart Failure With Reduced Ejection Fraction. JAMA Cardiol. August 2020. doi:10.1001/jamacardio.2020.2822 [ Links ]

20. López-Sendón J, Swedberg K, McMurray J, et al. Documento de Consenso de Expertos sobre el uso de inhibidores de la enzima de conversión de la angiotensina en la enfermedad cardiovascular. Rev Española Cardiol. 2004;57(12):1213-1232. doi:10.1157/13069868 [ Links ]

21. McMurray JJ, Ostergren J, Swedberg K, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors: the CHARM-Added trial. Lancet. 2003;362(9386):767-771. doi:10.1016/S0140-6736(03)14283-3 [ Links ]

22. Cohn JN, Tognoni G. A Randomized Trial of the Angiotensin-Receptor Blocker Valsartan in Chronic Heart Failure. N Engl J Med. 2001;345(23):1667-1675. doi:10.1056/NEJMoa010713 [ Links ]

23. Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomised trial-the Losartan Heart Failure Survival Study ELITE II. Lancet. 2000;355(9215):1582-1587. doi:10.1016/S0140-6736(00)02213-3 [ Links ]

24. Dickstein K, Kjekshus J. Effects of losartan and captopril on mortality and morbidity in high-risk patients after acute myocardial infarction: the OPTI-MAAL randomised trial. Lancet. 2002;360(9335):752-760. doi:10.1016/S0140-6736(02)09895-1 [ Links ]

25. Granger CB, McMurray JJ, Yusuf S, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function intolerant to angiotensin-converting-enzyme inhibitors: the CHARM-Alternative trial. Lancet. 2003;362(9386):772-776. doi:10.1016/S0140-6736(03)14284-5 [ Links ]

26. Foody JM, Farrell MH, Krumholz HM. p-Blocker Therapy in Heart Failure. JAMA. 2002;287(7):883. doi:10.1001/jama.287.7.883 [ Links ]

27. Shah NB, Der E, Ruggerio C, Heidenreich PA, Massie BM. Prevention of hospitalizations for heart failure with an interactive home monitoring program. Am Heart J. 1998;135(3):373-378. doi:10.1016/S0002-8703(98)70310-2 [ Links ]

28. Khand A, Gemmel I, Clark AL, Cleland JG. Is the prognosis of heart failure improving? J Am Coll Cardiol. 2000;36(7):2284-2286. doi:10.1016/S0735-1097(00)00995-5 [ Links ]

29. Bristow MR, Gilbert EM, Abraham WT, et al. Carvedilol Produces Dose-Related Improvements in Left Ventricular Function and Survival in Subjects With Chronic Heart Failure. Circulation. 1996;94(11):2807-2816. doi:10.1161/01.CIR.94.11.2807 [ Links ]

30. Murphy SP, Ibrahim NE, Januzzi JL. Heart Failure With Reduced Ejection Fraction. JAMA. 2020;324(5):488. doi:10.1001/jama.2020.10262 [ Links ]

31. Kotecha D, Holmes J, Krum H, et al. Efficacy of p blockers in patients with heart failure plus atrial fibrillation: an individual-patient data meta-analysis. Lancet. 2014;384(9961):2235-2243. doi:10.1016/S0140-6736(14)61373-8 [ Links ]

32. Kotecha D, Bunting K V., Gill SK, et al. Effect of Digoxin vs Bisoprolol for Heart Rate Control in Atrial Fibrillation on Patient-Reported Quality of Life. JAMA. 2020;324(24):2497. doi:10.1001/jama.2020.23138 [ Links ]

33. Struthers AD, Unger T. Physiology of aldosterone and pharmacology of aldosterone blockers. Eur Heart J Suppl B 2011; 13: 27B-30B. [ Links ]

34. Struthers AD. The clinical implications of aldosterone escape in congestive heart failure. Eur J Heart Fail. 2004;6(5):539-545. doi:10.1016/j.ejheart.2004.04.013 [ Links ]

35. Turban S, Wang X-Y, Knepper MA. Regulation of NHE3, NKCC2, and NCC abundance in kidney during aldosterone escape phenomenon: role of NO. Am J Physiol Physiol. 2003;285(5):F843-F851. doi:10.1152/ajprenal.00110.2003 [ Links ]

36. Zannad F, McMurray JJV, Krum H, et al. Eplerenone in Patients with Systolic Heart Failure and Mild Symptoms. N Engl J Med. 2011;364(1):11-21. doi:10.1056/NEJMoa1009492 [ Links ]

37. Pitt B, Remme W, Zannad F, et al. Eplerenone, a Selective Aldosterone Blocker, in Patients with Left Ventricular Dysfunction after Myocardial Infarction. N Engl J Med. 2003;348(14):1309-1321. doi:10.1056/NEJMoa030207 [ Links ]

38. Pitt B, Zannad F, Remme WJ, et al. The Effect of Spironolactone on Morbidity and Mortality in Patients with Severe Heart Failure. N Engl J Med. 1999;341(10):709-717. doi:10.1056/NEJM199909023411001 [ Links ]

39. Vaduganathan M, Claggett BL, Jhund PS, et al. Estimating lifetime benefits of comprehensive disease-modifying pharmacological therapies in patients with heart failure with reduced ejection fraction: a comparative analysis of three randomised controlled trials. Lancet. 2020;396(10244):121-128. doi:10.1016/S0140-6736(20)30748-0 [ Links ]

40. Filippatos G, Anker SD, Bohm M, et al. A randomized controlled study of finerenone vs. eplerenone in patients with worsening chronic heart failure and diabetes mellitus and/or chronic kidney disease. Eur Heart J. 2016;37(27):2105-2114. doi:10.1093/eurheartj/ehw132 [ Links ]

41. Bakris GL, Agarwal R, Anker SD, et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N Engl J Med. 2020;383(23):2219-2229. doi:10.1056/NEJMoa2025845 [ Links ]

42. Tsao CW, Lyass A, Enserro D, et al. Temporal Trends in the Incidence of and Mortality Associated With Heart Failure With Preserved and Reduced Ejection Fraction. JACC Hear Fail. 2018;6(8):678-685. doi:10.1016/j.jchf.2018.03.006 [ Links ]

43. Daniels LB, Maisel AS. Natriuretic Peptides. J Am Coll Cardiol. 2007;50(25):2357-2368. doi:10.1016/j.jacc.2007.09.021 [ Links ]

44. O'Connor CM, Starling RC, Hernandez AF, et al. Effect of Nesiritide in Patients with Acute Decompensated Heart Failure. N Engl J Med. 2011;365(1):32-43. doi:10.1056/NEJMoa1100171 [ Links ]

45. Hata N, Seino Y, Tsutamoto T, et al. Effects of Carperitide on the Long-Term Prognosis of Patients With Acute Decompensated Chronic Heart Failure. Circ J. 2008;72(11):1787-1793. doi:10.1253/circj.CJ-08-0130 [ Links ]

46. O.Connell JE, Jardine AG, Davidson G, Connell JMC. Candoxatril, an orally active neutral endopeptidase inhibitor, raises plasma atrial natriuretic factor and is natriuretic in essential hypertension. J Hypertens. 1992;10(3):271-277. doi:10.1097/00004872-199203000-00011 [ Links ]

47. Packer M, Califf RM, Konstam MA, et al. Comparison of Omapatrilat and Enalapril in Patients With Chronic Heart Failure. Circulation. 2002;106(8):920-926. doi:10.1161/01.CIR.0000029801.86489.50 [ Links ]

48. McMurray JJV, Packer M, Desai AS, et al. Angiotensin-Neprilysin Inhibition versus Enalapril in Heart Failure. N Engl J Med. 2014;371(11):993-1004. doi:10.1056/NEJMoa1409077 [ Links ]

49. Velazquez EJ, Morrow DA, DeVore AD, et al. Angiotensin-Neprilysin Inhibition in Acute Decompensated Heart Failure. N Engl J Med. 2019;380(6):539-548. doi:10.1056/NEJMoa1812851 [ Links ]

50. Senni M, McMurray JJV, Wachter R, et al. Initiating sacubitril/valsartan (LCZ696) in heart failure: results of TITRATION, a doubleDblind, randomized comparison of two uptitration regimens. Eur J Heart Fail. 2016; 18(9):1193-1202. doi:10.1002/ejhf.548 [ Links ]

51. Wachter R, Senni M, Belohlavek J, et al. Initiation of sacubitril/valsartan in haemodynamically stabilised heart failure patients in hospital or early after discharge: primary results of the randomised TRANSITION study. Eur J Heart Fail. 2019;21(8):998-1007. doi:10.1002/ejhf.1498 [ Links ]

52. Januzzi JL, Prescott MF, Butler J, et al. Association of Change in N-Terminal Pro-B-Type Natriuretic Peptide Following Initiation of Sacubitril-Valsartan Treatment With Cardiac Structure and Function in Patients With Heart Failure With Reduced Ejection Fraction. JAMA. 2019;322(11):1085. doi:10.1001/jama.2019.12821 [ Links ]

53. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373(22):2117-2128. doi:10.1056/NEJMoa1504720 [ Links ]

54. Hallow KM, Helmlinger G, Greasley PJ, McMurray JJ V., Boulton DW. Why do SGLT2 inhibitors reduce heart failure hospitalization? A differential volume regulation hypothesis. Diabetes, Obes Metab. 2018;20(3):479-487. doi:10.1111/dom.13126 [ Links ]

55. Matthews VB, Elliot RH, Rudnicka C, Hricova J, Herat L, Schlaich MP. Role of the sympathetic nervous system in regulation of the sodium glucose cotransporter 2. J Hypertens. 2017;35(10):2059-2068. doi:10.1097/HJH.0000000000001434 [ Links ]

56. Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, et al. Empagliflozin Ameliorates Adverse Left Ventricular Remodeling in Nondiabetic Heart Failure by Enhancing Myocardial Energetics. J Am Coll Cardiol. 2019;73(15):1931-1944. doi:10.1016/j.jacc.2019.01.056 [ Links ]

57. Nassif ME, Windsor SL, Tang F, et al. Dapagliflozin Effects on Biomarkers, Symptoms, and Functional Status in Patients With Heart Failure With Reduced Ejection Fraction. Circulation. 2019;140(18):1463-1476. doi:10.1161/CIRCU-LATIONAHA.119.042929 [ Links ]

58. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N Engl J Med. 2019;381(21):1995-2008. doi:10.1056/NEJMoa1911303 [ Links ]

59. Packer M, Anker SD, Butler J, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med. 2020;383(15):1413-1424. doi:10.1056/NEJMoa2022190 [ Links ]

60. Bhatt DL, Szarek M, Steg PG, et al. Sotagliflozin in Patients with Diabetes and Recent Worsening Heart Failure. N Engl J Med. November 2020:NEJ-Moa2030183. doi:10.1056/NEJMoa2030183 [ Links ]

61. Cannon CP, Pratley R, Dagogo-Jack S, et al. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N Engl J Med. 2020;383(15):1425-1435. doi:10.1056/NEJMoa2004967 [ Links ]

62. Cosentino F, Cannon CP, Cherney DZI, et al. Efficacy of Ertugliflozin on Heart Failure-Related Events in Patients With Type 2 Diabetes Mellitus and Established Atherosclerotic Cardiovascular Disease. Circulation. 2020;142(23):2205-2215. doi:10.1161/CIRCULATIONAHA.120.050255 [ Links ]

63. Fox K, Ford I, Steg PG, Tendera M, Ferrari R. Ivabradine for patients with stable coronary artery disease and left-ventricular systolic dysfunction (BEAUTIFUL): a randomised, double-blind, placebo-controlled trial. Lancet. 2008;372(9641):807-816. doi:10.1016/S0140-6736(08)61170-8 [ Links ]

64. Fox K, Ford I, Steg PG, Tendera M, Robertson M, Ferrari R. Heart rate as a prognostic risk factor in patients with coronary artery disease and left-ventricular systolic dysfunction (BEAUTIFUL): a subgroup analysis of a randomised controlled trial. Lancet. 2008;372(9641):817-821. doi:10.1016/S0140-6736(08)61171-X [ Links ]

65. Swedberg K, Komajda M, Bohm M, et al. Ivabradine and outcomes in chronic heart failure (SHIFT): a randomised placebo-controlled study. Lancet. 2010;376(9744):875-885. doi:10.1016/S0140-6736(10)61198-1 [ Links ]

66. Kotecha D, Flather MD, Altman DG, et al. Heart Rate and Rhythm and the Benefit of Beta-Blockers in Patients With Heart Failure. J Am Coll Cardiol. 2017;69(24):2885-2896. doi:10.1016/j.jacc.2017.04.001 [ Links ]

67. Cullington D, Goode KM, Cleland JGF, Clark AL. Limited role for ivabradine in the treatment of chronic heart failure. Heart. 2011;97(23):1961-1966. doi:10.1136/heartjnl-2011-300041 [ Links ]

68. Cohn JN, Archibald DG, Ziesche S, et al. Effect of Vasodilator Therapy on Mortality in Chronic Congestive Heart Failure. N Engl J Med. 1986;314(24):1547-1552. doi:10.1056/NEJM198606123142404 [ Links ]

69. Cohn JN, Johnson G, Ziesche S, et al. A Comparison of Enalapril with Hydralazine-Isosorbide Dinitrate in the Treatment of Chronic Congestive Heart Failure. N Engl J Med. 1991;325(5):303-310. doi:10.1056/NEJM199108013250502 [ Links ]

70. Taylor AL, Ziesche S, Yancy C, et al. Combination of Isosorbide Dinitrate and Hydralazine in Blacks with Heart Failure. N Engl J Med. 2004;351(20):2049-2057. doi:10.1056/NEJMoa042934 [ Links ]

71. Gheorghiade M, Greene SJ, Butler J, et al. Effect of Vericiguat, a Soluble Guanylate Cyclase Stimulator, on Natriuretic Peptide Levels in Patients With Worsening Chronic Heart Failure and Reduced Ejection Fraction. JAMA. 2015;314(21):2251. doi:10.1001/jama.2015.15734 [ Links ]

72. Armstrong PW, Pieske B, Anstrom KJ, et al. Vericiguat in Patients with Heart Failure and Reduced Ejection Fraction. N Engl J Med. 2020;382(20):1883-1893. doi:10.1056/NEJMoa1915928 [ Links ]

73. Teerlink JR, Diaz R, Felker GM, et al. Cardiac Myosin Activation with Omecamtiv Mecarbil in Systolic Heart Failure. N Engl J Med. November 2020:NEJMoa2025797. doi:10.1056/NEJMoa2025797 [ Links ]

74. Gheorghiade M, Adams KF, Colucci WS. Digoxin in the Management of Cardiovascular Disorders. Circulation. 2004;109(24):2959-2964. doi:10.1161/01.CIR.0000132482.95686.87 [ Links ]

75. The Effect of Digoxin on Mortality and Morbidity in Patients with Heart Failure. N Engl J Med. 1997;336(8):525-533. doi:10.1056/NEJM199702203360801 [ Links ]

76. Gheorghiade M, Patel K, Filippatos G, et al. Effect of oral digoxin in high-risk heart failure patients: a pre-specified subgroup analysis of the DIG trial. Eur J Heart Fail. 2013;15(5):551-559. doi:10.1093/eurjhf/hft010 [ Links ]

77. Hood Jr. WB, Dans AL, Guyatt GH, Jaeschke R, McMurray JJ. Digitalis for treatment of heart failure in patients in sinus rhythm. Cochrane Database Syst Rev. April 2014. doi:10.1002/14651858.CD002901.pub3 [ Links ]

78. Patterson JH, Adams KF, Applefeld MM, Corder CN, Masse BR. Oral Torsemide in Patients With Chronic Congestive Heart Failure: Effects on Body Weight, Edema, and Electrolyte Excretion. Pharmacother J Hum Pharmacol Drug Ther. 1994;14(5):514-521. doi:10.1002/j.1875-9114.1994.tb02848.x [ Links ]

79. Anand IS, Kalra GS, Harris P et al. Diuretics as initial and sole treatment in chronic cardiac failure. Cardioscience. 2(4):273-8. [ Links ]

80. Faris RF, Flather M, Purcell H, Poole-Wilson PA, Coats AJ. Diuretics for heart failure. Cochrane Database Syst Rev. April 2016. doi:10.1002/14651858.CD003838.pub4 [ Links ]

81. Gattis W a, Connor CMO, Adams KF, et al. Effects of Tolvaptan Heart Fail. 2004;291(16):1963-1971. [ Links ]

82. Konstam MA. Effects of Oral Tolvaptan in Patients Hospitalized for Worsening Heart Failure The EVEREST Outcome Trial. JAMA. 2007;297(12):1319. doi:10.1001/jama.297.12.1319 [ Links ]

83. Jankowska EA, Rozentryt P, Witkowska A, et al. Iron deficiency: an ominous sign in patients with systolic chronic heart failure. Eur Heart J. 2010;31(15):1872-1880. doi:10.1093/eurheartj/ehq158 [ Links ]

84. Comín-Colet J, Enjuanes C, González G, et al. Iron deficiency is a key determinant of health-related quality of life in patients with chronic heart failure regardless of anaemia status. Eur J Heart Fail. 2013;15(10):1164-1172. doi:10.1093/eurjhf/hft083 [ Links ]

85. Enjuanes C, Klip IjT, Bruguera J, et al. Iron deficiency and health-related quality of life in chronic heart failure: Results from a multicenter European study. Int J Cardiol. 2014;174(2):268-275. doi:10.1016/j.ijcard.2014.03.169 [ Links ]

86. Klip IjT, Comin-Colet J, Voors AA, et al. Iron deficiency in chronic heart failure: An international pooled analysis. Am Heart J. 2013;165(4):575-582.e3. doi:10.1016/j.ahj.2013.01.017 [ Links ]

87. Ponikowski P, van Veldhuisen DJ, Comin-Colet J, et al. Beneficial effects of long-term intravenous iron therapy with ferric carboxymaltose in patients with symptomatic heart failure and iron deficiency. Eur Heart J. 2015;36(11):657-668. doi:10.1093/eurheartj/ehu385 [ Links ]

88. Anker SD, Comin Colet J, Filippatos G, et al. Ferric Carboxymaltose in Patients with Heart Failure and Iron Deficiency. N Engl J Med. 2009;361(25):2436-2448. doi:10.1056/NEJMoa0908355 [ Links ]

89. Ponikowski P, Kirwan B-A, Anker SD, et al. Ferric carboxymaltose for iron deficiency at discharge after acute heart failure: a multicentre, double-blind, randomised, controlled trial. Lancet. 2020;396(10266):1895-1904. doi:10.1016/S0140-6736(20)32339-4 [ Links ]

90. Lewis GD, Malhotra R, Hernandez AF, et al. Effect of Oral Iron Repletion on Exercise Capacity in Patients With Heart Failure With Reduced Ejection Fraction and Iron Deficiency. JAMA. 2017;317(19):1958. doi:10.1001/jama.2017.5427 [ Links ]

91. Maisel WH, Stevenson LW. Atrial fibrillation in heart failure: epidemiology, pathophysiology, and rationale for therapy. Am J Cardiol. 2003;91(6):2-8. doi:10.1016/S0002-9149(02)03373-8 [ Links ]

92. Mamas MA, Caldwell JC, Chacko S, Garratt CJ, Fath-Ordoubadi F, Neyses L. A meta-analysis of the prognostic significance of atrial fibrillation in chronic heart failure. Eur J Heart Fail. 2009;11(7):676-683. doi:10.1093/eurjhf/hfp085 [ Links ]

93. Agarwal M, Apostolakis S, Lane DA, Lip GYH. The Impact of Heart Failure and Left Ventricular Dysfunction in Predicting Stroke, Thromboembolism, and Mortality in Atrial Fibrillation Patients: A Systematic Review. Clin Ther. 2014;36(9):1135-1144. doi:10.1016/j.clinthera.2014.07.015 [ Links ]

94. Hindricks G, Potpara T, Dagres N, et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J. August 2020. doi:10.1093/eurheartj/ehaa612 [ Links ]

95. Cadrin-Tourigny J, Shohoudi A, Roy D, et al. Decreased Mortality With Beta-Blockers in Patients With Heart Failure and Coexisting Atrial Fibrillation. JACC Hear Fail. 2017;5(2):99-106. doi:10.1016/j.jchf.2016.10.015 [ Links ]

96. Marrouche NF, Brachmann J, Andresen D, et al. Catheter Ablation for Atrial Fibrillation with Heart Failure. N Engl J Med. 2018;378(5):417-427. doi:10.1056/NEJMoa1707855 [ Links ]

97. MERIT-HF Study Group. Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in-Congestive Heart Failure (MERIT-HF). Lancet. 1999;353(9169):2001-2007. doi:10.1016/S0140-6736(99)04440-2 [ Links ]

98. Al-Khatib SM, Stevenson WG, Ackerman MJ, et al. 2017 AHA/ACC/HRS Guideline for Management of Patients With Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death. J Am Coll Cardiol. 2018;72(14):e91-e220. doi:10.1016/j.jacc.2017.10.054 [ Links ]

99. Moss AJ, Hall WJ, Cannom DS, et al. Improved Survival with an Implanted Defibrillator in Patients with Coronary Disease at High Risk for Ventricular Arrhythmia. N Engl J Med. 1996;335(26):1933-1940. doi:10.1056/NEJM199612263352601 [ Links ]

100. Moss AJ, Zareba W, Hall WJ, et al. Prophylactic Implantation of a Defibrillator in Patients with Myocardial Infarction and Reduced Ejection Fraction. N Engl J Med. 2002;346(12):877-883. doi:10.1056/NEJMoa013474 [ Links ]

101. Al-Khatib SM, Fonarow GC, Joglar JA, et al. Primary Prevention Implant-able Cardioverter Defibrillators in Patients With Nonischemic Cardiomyopathy. JAMA Cardiol. 2017;2(6):685. doi:10.1001/jamacardio.2017.0630 [ Links ]

102. Køber L, Thune JJ, Nielsen JC, et al. Defibrillator Implantation in Patients with Nonischemic Systolic Heart Failure. N Engl J Med. 2016;375(13):1221-1230. doi:10.1056/NEJMoa1608029 [ Links ]

103. Khan NK, Goode KM, Cleland JGF, et al. Prevalence of ECG abnormalities in an international survey of patients with suspected or confirmed heart failure at death or discharge. Eur J Heart Fail. 2007;9(5):491-501. doi:10.1016/j.ejheart.2006.11.003 [ Links ]

104. Cleland JGF, Daubert J-C, Erdmann E, et al. The Effect of Cardiac Resynchronization on Morbidity and Mortality in Heart Failure. N Engl J Med. 2005;352(15):1539-1549. doi:10.1056/NEJMoa050496 [ Links ]

105. Bristow MR, Saxon LA, Boehmer J, et al. Cardiac-Resynchronization Therapy with or without an Implantable Defibrillator in Advanced Chronic Heart Failure. N Engl J Med. 2004;350(21):2140-2150. doi:10.1056/NEJMoa032423 [ Links ]

106. Moss AJ, Hall WJ, Cannom DS, et al. Cardiac-Resynchronization Therapy for the Prevention of Heart-Failure Events. N Engl J Med. 2009;361(14):1329-1338. doi:10.1056/NEJMoa0906431 [ Links ]

107. Borggrefe MM, Lawo T, Butter C, et al. Randomized, double blind study of non-excitatory, cardiac contractility modulation electrical impulses for symptomatic heart failure. Eur Heart J. 2008;29(8):1019-1028. doi:10.1093/eurheartj/ehn020 [ Links ]

108. Neelagaru SB, Sanchez JE, Lau SK, et al. Nonexcitatory, cardiac contractility modulation electrical impulses: Feasibility study for advanced heart failure in patients with normal QRS duration. Hear Rhythm. 2006;3(10):1140-1147. doi:10.1016/j.hrthm.2006.06.031 [ Links ]

109. Kadish A, Nademanee K, Volosin K, et al. A randomized controlled trial evaluating the safety and efficacy of cardiac contractility modulation in advanced heart failure. Am Heart J. 2011;161(2):329-337.e2. doi:10.1016/j.ahj.2010.10.025 [ Links ]

110. Abraham WT, Nademanee K, Volosin K, et al. Subgroup Analysis of a Randomized Controlled Trial Evaluating the Safety and Efficacy of Cardiac Contractility Modulation in Advanced Heart Failure. J Card Fail. 2011;17(9):710-717. doi:10.1016/j.cardfail.2011.05.006 [ Links ]

111. Abraham WT, Kuck K-H, Goldsmith RL, et al. A Randomized Controlled Trial to Evaluate the Safety and Efficacy of Cardiac Contractility Modulation. JACC Hear Fail. 2018;6(10):874-883. doi:10.1016/j.jchf.2018.04.010 [ Links ]

112. Grigioni F, Enriquez-Sarano M, Zehr KJ, Bailey KR, Tajik AJ. Ischemic Mitral Regurgitation. Circulation. 2001;103(13):1759-1764. doi:10.1161/01.CIR.103.13.1759 [ Links ]

113. Goliasch G, Bartko PE, Pavo N, et al. Refining the prognostic impact of functional mitral regurgitation in chronic heart failure. Eur Heart J. 2018;39(1):39-46. doi:10.1093/eurheartj/ehx402 [ Links ]

114. Mirabel M, Iung B, Baron G, et al. What are the characteristics of patients with severe, symptomatic, mitral regurgitation who are denied surgery? Eur Heart J. 2007;28(11):1358-1365. doi:10.1093/eurheartj/ehm001 [ Links ]

115. Obadia J-F, Messika-Zeitoun D, Leurent G, et al. Percutaneous Repair or Medical Treatment for Secondary Mitral Regurgitation. N Engl J Med. 2018;379(24):2297-2306. doi:10.1056/NEJMoa1805374 [ Links ]

116. Stone GW, Lindenfeld J, Abraham WT, et al. Transcatheter Mitral-Valve Repair in Patients with Heart Failure. N Engl J Med. 2018;379(24):2307-2318. doi:10.1056/NEJMoa1806640 [ Links ]

117. Pibarot P, Delgado V, Bax JJ. MITRA-FR vs. COAPT: lessons from two trials with diametrically opposed results. Eur Hear J-Cardiovasc Imaging. 2019;20(6):620-624. doi:10.1093/ehjci/jez073 [ Links ]

118. Grayburn PA, Sannino A, Packer M. Proportionate and Disproportionate Functional Mitral Regurgitation: A New Conceptual Framework That Reconciles the Results of the MITRA-FR and COAPT Trials. JACC Cardiovasc Imaging. 2019;12(2):353-362. doi:10.1016/j.jcmg.2018.11.006 [ Links ]

119. Hagendorff A, Knebel F, Helfen A, Stobe S, Doenst T, Falk V. Disproportionate mitral regurgitation: another myth? A critical appraisal of echocardiographic assessment of functional mitral regurgitation. Int J Cardiovasc Imaging. August 2020. doi:10.1007/s10554-020-01975-6 [ Links ]

120. Otto CM, Nishimura RA, Bonow RO, et al. 2020 ACC/AHA Guideline for the Management of Patients With Valvular Heart Disease: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. December 2020. doi:10.1161/CIR.0000000000000923 [ Links ]

121. Angermann CE, Assmus B, Anker SD, et al. Pulmonary artery pressure D guided therapy in ambulatory patients with symptomatic heart failure: the Cardio MEMS European Monitoring Study for Heart Failure (MEMSDHF). Eur J Heart Fail. 2020;22(10):1891-1901. doi:10.1002/ejhf.1943 [ Links ]

122. Shavelle DM, Desai AS, Abraham WT, et al. Lower Rates of Heart Failure and All-Cause Hospitalizations During Pulmonary Artery Pressure-Guided Therapy for Ambulatory Heart Failure. Circ Hear Fail. 2020;13(8). doi:10.1161/CIRCHEARTFAILURE.119.006863 [ Links ]

123. Givertz MM, Stevenson LW, Costanzo MR, et al. Pulmonary Artery Pressure-Guided Management of Patients With Heart Failure and Reduced Ejection Fraction. J Am Coll Cardiol. 2017;70(15):1875-1886. doi:10.1016/j.jacc.2017.08.010 [ Links ]

124. Dhruva SS, Krumholz HM. Championing Effectiveness Before Cost-Effectiveness D. JACC Hear Fail. 2016;4(5):376-379. doi: 10.1016/j.jchf.2016.02.001 [ Links ]

125. Rose EA, Gelijns AC, Moskowitz AJ, et al. Long-Term Use of a Left Ventricular Assist Device for End-Stage Heart Failure. N Engl J Med. 2001;345(20):1435-1443. doi:10.1056/NEJMoa012175 [ Links ]

126. Aaronson KD, Slaughter MS, Miller LW, et al. Use of an Intrapericardial, Continuous-Flow, Centrifugal Pump in Patients Awaiting Heart Transplantation. Circulation. 2012;125(25):3191-3200. doi:10.1161/CIRCULATIONAHA.111.058412 [ Links ]

127. Rogers JG, Pagani FD, Tatooles AJ, et al. Intrapericardial Left Ventricular Assist Device for Advanced Heart Failure. N Engl J Med. 2017;376(5):451-460. doi:10.1056/NEJMoa1602954 [ Links ]

128. Mehra MR, Uriel N, Naka Y, et al. A Fully Magnetically Levitated Left Ventricular Assist Device - Final Report. N Engl J Med. 2019;380(17):1618-1627. doi:10.1056/NEJMoa1900486 [ Links ]

129. Potapov EV, Antonides C, Crespo-Leiro MG, et al. 2019 EACTS Expert Consensus on long-term mechanical circulatory support. Eur J Cardio-Thoracic Surg. 2019;56(2):230-270. doi:10.1093/ejcts/ezz098 [ Links ]

130. Docherty KF, Campbell RT, Jhund PS, Petrie MC, McMurray JJV. How robust are clinical trials in heart failure? Eur Heart J. October 2016:ehw427. doi:10.1093/eurheartj/ehw427 [ Links ]

131. Greene SJ, Butler J, Fonarow G. Simultaneous or Rapid Sequence Initiation of Quadruple Medical Therapy for Heart Failure- Optimizing Therapy With the Need for Speed. JAMA Cardiol 2021 Mar 31. doi: 10.1001/jamacardio.2021.0496 [ Links ]

132. Mehra MR, Canter CE, Hannan MM, et al. The 2016 International Society for Heart Lung Transplantation listing criteria for heart transplantation: A 10-year update. J Hear Lung Transplant. 2016;35(1):1-23. doi:10.1016/j.healun.2015.10.023 [ Links ]

Anexo

Tratamiento de comorbilidades

Suplementación de hierro

La deficiencia de hierro está estrechamente relacionada con la gravedad de la FC y se asocia a un mayor riesgo de muerte, independientemente de la presencia de anemia 83, peor clase funcional, pobre capacidad máxima al esfuerzo y peor calidad de vida 84,85. La deficiencia de hierro afecta al 50% de los pacientes con FC 86.

La administración intravenosa de hierro se ha estudiado específicamente en dos estudios aleatorizados (FAIR-HF y CONFIRM-HF) 87,88 en pacientes con FC y déficit de hierro demostraron que el hierro carboximaltosa intravenoso mejora el estado general de los pacientes, la calidad de vida y la clase funcional de la NYHA. Como objetivo secundario, se observó una reducción significativa de la hospitalización por empeoramiento de la FC. El reciente ensayo AFFIRM AHF en 1132 pacientes con hospitalización por FC aguda y asociada a deficiencia de hierro, demostró impacto sobre el desenlace combinado de hospitalizaciones totales por FC y muerte de origen cardiovascular, aunque casi exclusivamente a expensas de la reducción en las hospitalizaciones (57.2 pacientes/año vs 72.5 pacientes/año) 89,90.

Fibrilación auricular

Se reporta una prevalencia de FA del 4.2% en pacientes NYHA I y hasta el 49.8% en pacientes NYHA IV 91. El desarrollo de FA en esta población se asocia a un peor pronóstico, asociándose con un aumento significativo de la mortalidad global (OR 1.4, IC 1.32-1.48) 92; en todos los pacientes con disfunción ventricular y FA está indicada la anticoagulación dado que la presencia de FC confiere un aumento de 1.7-2.6 veces en el riesgo fenómenos embólicos 93. Para el control de la respuesta ventricular se pueden emplear betabloqueadores, digoxina y como último recurso amiodarona. Los calcioantagonistas no dihidropiridínicos están contraindicados. A pesar de que no ha sido estudiado específicamente, en estos pacientes se propone una meta de frecuencia cardiaca entre < 100-110 latidos por minuto 94.

Aunque ciertos betabloqueadores tienen claro beneficio sobre la mortalidad en FC-FEVIr, el beneficio de esta terapia parece atenuarse en los pacientes que de manera concomitante presentan FA. Así, estudios muestran que los betabloqueadores disminuyen la mortalidad del paciente en ritmo sinusal (HR 0.73, IC 0.67-0.8) pero no en ritmo de FA (0.97, IC 0.83-1.14). A pesar de esto, los betabloqueadores son seguros y algunos estudios sugieren que podría haber un efecto benéfico por lo que se recomienda su uso 95; como alternativa puede utilizarse la digoxina con menores efectos secundarios de acuerdo con los resultados del ensayo RATE-AF 32.

El aislamiento eléctrico de venas pulmonares es una alternativa atractiva; el estudio CASTLE-AF encontró que los pacientes con FEVI < 35% y FA con pobre respuesta al tratamiento que se asignaron a ablación por catéter tuvieron una disminución en el desenlace combinado de muerte u hospitalización por FC al compararse con la terapia estándar (HR 0.62, IC 0.43-0.87) 96; sin embargo, el efecto solo pudo ser manifiesto después de dos años de seguimiento. Estudios adicionales en esta área son necesarios para definir el papel definitivo de esta terapia.

Tratamiento con dispositivos

Desfibrilador automático implantable (DAI)

La causa de muerte en el paciente con FC varía según la clase funcional 97. En NYHA II, 64% de las muertes son de origen súbito y 12% por progresión de la FC; mientras que la mayoría de las muertes en NYHA IV son secundarias a progresión de la enfermedad (56%). Así, la mayoría de los estudios evaluando el papel del DAI han incluido pacientes sintomáticos NYHA II/III. En NYHA IV se desaconseja el uso de esta terapia (recomendación clase III) dado que esta población fue excluida de la mayoría de los estudios y que su pronóstico es pobre sin trasplante cardiaco 98.

El uso del DAI está recomendado para prevención primaria en pacientes con FEVI <35% luego de tres meses de tratamiento cuando se tiene una expectativa de vida superior a un año. El MADIT I fue el primer estudio en evaluar esta intervención. En éste, pacientes con cardiopatía isquémica, FEVI <35% y episodios de taquicardia ventricular no sostenida, tuvieron una reducción en la mortalidad con el uso de DAI (HR 0.46; IC 0.26-0.82) 99. En el MADIT II, los pacientes con cardiopatía isquémica y FEVI <30% que fueron asignados a DAI, tuvieron una menor mortalidad global a dos años (14.2% vs 19.8%; HR 0,69, IC 0.51-0.93) 100.

El uso de DAI en cardiopatía no isquémica tiene una evidencia menos sólida, aunque hay estudios que avalan su uso 101. El 50% de la población incluida en el SCD-HeFT tenía una cardiopatía no isquémica. En este ensayo, pacientes con FEVI <35% y clase funcional NYHA II/III se asignaron a placebo, amiodarona o DAI. El uso de DAI se asoció a una reducción en el riesgo de muerte (HR 0.77, IC 0.62-0.96) sin haber diferencias en el subanálisis según la etiología de la FC.

Por otro lado, el estudio DANISH incluyó paciente específicamente de origen no isquémico. Aunque en el estudio, hubo diferencias en el desenlace secundario de muerte súbita (HR 0.5, IC 0.31-0.82), no hubo diferencias en el desenlace primario de mortalidad (HR 0.87, IC 0.681.12) 102.

Terapia de resincronización cardiaca (TRC)

La disincronía ventricular es un hallazgo frecuente en los pacientes con FC. Aunque existen diferentes maneras para estimar la misma, la duración del QRS sigue siendo el método más adecuado. Un QRS prolongado (>120 milisegundos) es un hallazgo frecuente en el paciente con FC (28% de los casos) 103. La TRC busca mejorar la disincronía electromecánica mediante la estimulación biventricular e impacta de manera favorable la FEVI, el remodelamiento cardiaco, el grado de insuficiencia mitral y los niveles de NTproBNP (104). Además, algunos estudios han demostrado beneficio en desenlaces de mortalidad y hospitalización por FC 104-106.

La FEVI, la duración del QRS, la morfología del bloqueo de rama y la clase funcional son los parámetros empleados para definir la necesidad de TRC. El COMPANION, evaluó la utilidad de la TRC en pacientes NYHA III/IV, con FEVI <35% y QRS >120 milisegundos. Este estudio comparó de manera aleatoria los grupos de manejo farmacológico, TRC, o TRC con DAI. El uso de TRC disminuyó de manera significativa el desenlace combinado de muerte y hospitalización (HR 0.81; IC 0.69-0.96). En el análisis por subgrupos, no hubo beneficio de la intervención en presencia de bloqueos diferentes a rama izquierda 105.

El estudio MADIT-CRT incluyó pacientes con FEVI <30%, QRS >130 milisegundos y una clase funcional NYHA I/II. Los pacientes se asignaron a TRC con DAI vs DAI de manera aislada. El ensayo encontró que los pacientes asignados a TRC con DAI tenían un menor desenlace combinado de muerte o eventos asociados a FC (17.2% vs 25.3%, HR 0.66; IC 0.52-0.84). En el análisis por subgrupos se encontró que las personas con QRS <150 milisegundos no tenían un beneficio de la intervención (HR 1.06, IC 0.74-1.52) 106.

Modulación de la contractilidad cardiaca

La modulación de la contractilidad cardiaca consiste en la estimulación eléctrica con señal bifásica, de alto voltaje y larga duración de la pared septal del ventrículo derecho durante el periodo refractario absoluto; la señal no produce una nueva contracción, sino que influencia la biología del miocardio en falla. Esto resulta en un incremento de la contractilidad cardiaca sin aumentar el consumo de oxígeno y genera remodelado reverso. Después de completar con éxito un estudio doble ciego y doble cruzado en Europa (FIX-HF-4) 107 y un estudio piloto en los Estados Unidos 108 se diseñó el estudio FIX-HF-5 para estudiar prospectivamente la seguridad y eficacia de la modulación de la contractilidad cardiaca en pacientes con FC con síntomas NYHA III o IV y FEVI <35% 109. Se incluyeron 428 pacientes, alcanzando el desenlace primario de seguridad (una evaluación de no inferioridad de la combinación de mortalidad por todas las causas y hospitalizaciones por todas las causas). Sin embargo, el desenlace primario de eficacia, que consistía en un análisis de los respondedores de los cambios en el umbral anaeróbico ventilatorio en las pruebas de esfuerzo cardiopulmonar, no se cumplió. Un análisis de subgrupos preespecificado mostró efectos significativos del tratamiento en los desenlaces primarios y secundarios (incluido el VO2 pico y el cuestionario Minnesota Living With Heart Failure Questionnaire) en pacientes con FEVI que oscilaban entre el 25% y el 45% 110. Con base en estos hallazgos se diseñó el estudio confirmatorio FIX-HF-5C 111, con el fin de confirmar prospectivamente la eficacia de la modulación de contractilidad cardiaca en pacientes con FEVI 25%-45%, encontrando mejoría en la tolerancia al ejercicio y la calidad de vida en el grupo especificado de pacientes con FC, y menores hospitalizaciones por FC.

Clip mitral

Se sabe que en los pacientes con FC e insuficiencia mitral secundaria hasta el 62% habrán muerto a cinco años 112-114. En este escenario surge, como una modificación a la técnica ideada por Alfieri en 1991, el reparo borde a borde por método percutáneo, más conocido como Mitra-Clip®, con el objetivo de obtener las presuntas ventajas de la reducción de la insuficiencia mitral funcional, mediante un método menos invasivo como es la implantación con catéter percutáneo.

Los estudios clínicos en este escenario han sido contradictorios así en el estudio MITRA-FR 115 en pacientes con FEVI 15-40% y NYHA ≥II a pesar de tratamiento óptimo con insuficiencia mitral secundaria con volumen regurgitante ≥30 cc u orificio regurgitante ≥20 mm2, los cuales no fuesen candidatos quirúrgicos no se logró demostrar la superioridad del MitraClip® aunado al manejo médico frente al manejo médico exclusivo en disminución de hospitalización por FC o muerte cardiovascular; en contraposición, en el estudio COAPT 116 pacientes con FEVI 20-50% y NYHA ≥II a pesar de tratamiento óptimo con insuficiencia mitral secundaria clasificada como 3+ o 4+, los cuales no fuesen candidatos quirúrgicos se demostró la superioridad del MitraClip® aunado al manejo médico frente al manejo médico exclusivo en disminución de hospitalización por FC (HR 0.53 IC 0.40- 0.70) o muerte (HR 0.62 IC 0.46-0.82).

Estos hallazgos generaron el concepto de regurgitación mitral proporcionada y desproporcionada, en el cual se plantea que existen pacientes cuya regurgitación mitral secundaria es proporcional al grado de dilatación ventricular por lo que en teoría responderían a terapias para reducir el volumen de fin de diástole ventricular izquierdo, en contraposición a otro grupo cuya severidad de la regurgitación es desproporcionada con respecto al volumen ventricular, por lo que se beneficiarían de intervenciones dirigidas a la válvula mitral 117-118; la propuesta es bastante seductora y ha sido aceptada hasta ahora, aunque sigue generando controversia 119, por lo que es importante estar atentos a nueva evidencia.

Por lo anteriormente expuesto, se puede considerar que en aquellos pacientes con FEVI 20-50% con un diámetro de fin de diástole del ventrículo izquierdo ≤ 70 mm con presión arterial pulmonar ≤ 70 mmHg, que persistan en NYHA ≥II a pesar de tratamiento óptimo y que tengan regurgitación mitral secundaria severa, el uso de MitraClip® estaría indicado (120).

CardioMEMS

La vigilancia remota de las variables hemodinámicas es un campo actual de investigación, de especial interés entre los pacientes con síntomas graves o FC avanzada. De forma convencional los DAI y TRC proporcionan información y alertas remotas que conducen a ajustes en el tratamiento del paciente. El dispositivo CardioMEMS, mide variables hemodinámicas en la arteria pulmonar y envía información de manera remota, su uso en pacientes con FC clase funcional NYHA III, se asoció con una reducción en el riesgo de hospitalizaciones por FC (HR 0.72 IC95% 0.59-0.88), con una presentación de complicaciones que no superaba el 1%, resultados reproducidos en el estudio MEMES HF 121, que se mantienen a un año de seguimiento 122. A pesar de tener aprobación por entidades regulatorias, las recomendaciones de las sociedades científicas son débiles dada la incertidumbre generada por el diseño de los estudios mencionados y por su costo-efectividad 123,124.

Asistencia ventricular

El estudio clásico fue el REMATCH publicado en 2001 incluyó pacientes con FC avanzada con contraindicación para trasplante, el cual demostró reducción de mortalidad comparado con placebo (RR 0.52 IC 95% 0.34-0.78) 125. Los desarrollos posteriores se centraron en mejorar el tamaño, la biocompatibilidad, durabilidad y disminuir la tasa de infecciones, pasando por dispositivos de segunda generación como el Heart Mate II, hasta llegar a los actuales, de tercera generación, con flujo continuo generado por bombas centrifugas como el Heart Ware y el Heart Mate III.

El ADVANCE trial con el dispositivo Heart Ware demostró supervivencia de 86% al año de seguimiento 126; posteriormente el ENDURANCE Trial demostró no inferioridad comparado con otros dispositivos de asistencia ventricular, con menor tasa de disfunción o falla del dispositivo, pero con aumento de la tasa de eventos cerebrovasculares y similar ganancia en calidad de vida 127. Finalmente, el MOMENTUM 3 Trial con el dispositivo Heart Mate 3 demostró superioridad con respecto a los dispositivos convencionales en cuanto a supervivencia y libre de enfermedad cerebrovascular o necesidad de reemplazo o remoción por disfunción a los dos años de seguimiento (RR 0.84 IC 95% 0.78-0.91) 128.

Por lo anterior el soporte mecánico circulatorio a largo plazo debe ser considerado en pacientes con estado funcional NYHA III-IV a pesar de manejo médico óptimo, con FEVI ≤ 25% y al menos otro criterio como: clasificación INTERMACS 2-4, dependencia de inotrópicos, disfunción de órgano progresiva, VO2 pico < 12 ml/Kg/minuto o dependencia de soporte mecánico temporal 129.

Received: February 08, 2021; Accepted: August 20, 2021

*Correspondencia: Dr. Juan Manuel Senior-Sánchez. Medellín (Colombia). E-Mail: juan.senior64@gmail.com

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License