Review Article

Optimizing Guideline-directed Medical Therapies for Heart Failure with Reduced Ejection Fraction During Hospitalization

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

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

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

Abstract

Heart failure remains a huge societal concern despite medical advancement, with an annual direct cost of over $30 billion. While guideline-directed medical therapy (GDMT) is proven to reduce morbidity and mortality, many eligible patients with heart failure with reduced ejection fraction (HFrEF) are not receiving one or more of the recommended medications, often due to suboptimal initiation and titration in the outpatient setting. Hospitalization serves as a key point to initiate and titrate GDMT. Four evidence-based therapies have clinical benefit within 30 days of initiation and form a crucial foundation for HFrEF therapy: renin-angiotensin-aldosterone system inhibitors with or without a neprilysin inhibitor, β-blockers, mineralocorticoid-receptor-antagonists, and sodium-glucose cotransporter-2 inhibitors. The authors present a practical guide for the implementation of these four pillars of GDMT during a hospitalization for acute heart failure.

Disclosure:GF has consulted for Abbott, Amgen, Bayer, Janssen, Medtronic, Merck, and Novartis. All other authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Jeffrey J Hsu, UCLA Center for Health Sciences, A2-237, 650 Charles E Young Drive South, Los Angeles, CA 90095-1679. E: jjhsu@mednet.ucla.edu

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

In the US, the annual direct cost of heart failure (HF) is estimated at over $30 billion, largely driven by the >1 million hospitalizations in which HF is the primary discharge diagnosis.1–3 The subset of patients with HF with reduced ejection fraction (HFrEF) comprises about half of all patients with HF.4 Guideline-directed medical therapy (GDMT) is proven to reduce mortality and morbidity for patients with HFrEF. GDMT includes the following drug therapies: renin-angiotensin-aldosterone system inhibitors (RAAS-I), with or without a neprilysin inhibitor, β-blockers, and mineralocorticoid-receptor-antagonists (MRA).5

Recently, sodium-glucose cotransporter-2 inhibitors (SGLT2i) demonstrated efficacy as an important fourth pillar of GDMT.6 Together, this combination can add over six additional years of lifespan for HFrEF patients compared to the traditional approach of RAAS-I and β-blockers alone.7 However, studies highlight that many eligible HFrEF patients are not receiving one or more of the recommended medications, in the absence of contraindications or intolerance.8 Even among patients who are treated, less than half receive optimal doses of GDMT. Additionally, time to initiation and optimization of dosing may be exceedingly slow in the outpatient setting.

Analysis of the CHAMP registry showed that less than 1% of patients receive target doses of RAAS-I/angiotensin receptor–neprilysin inhibitor (ARNI), β-blockers, and MRA simultaneously over a 12-month period.8,9 Guideline recommendations for device therapy, such as ICD and cardiac resynchronization therapy (CRT), recommend consideration only after 3 months of GDMT optimization. Yet, target doses of GDMT are rarely achieved prior to device therapy implantation.5,10 Barriers to optimization include lack of access and coordination of care, gaps in provider knowledge, and patient-related factors such as renal function, perceived intolerance to medications, inadequate insurance coverage, out-of-pocket expenses, and adherence.11,12 Hospitalization provides an opportunity to initiate and titrate medical therapy with close monitoring in patients with new-onset or acute on chronic HFrEF.13 Medications provided at discharge are more likely to be adhered to, continued, and further titrated in the outpatient setting.14 An acute heart failure (AHF) hospitalization is an important opportunity to initiate and titrate all four pillars of GDMT (Figure 1), laying a complete foundation for further outpatient optimization. We present a practical guide for the initiation and titration of evidence-based chronic HFrEF therapies during hospitalization.

Renin-Angiotensin-Aldosterone-Inhibitor/Angiotensin Receptor–Neprilysin Inhibitor

According to expert consensus, optimization of GDMT in an AHF hospitalization should occur once a positive clinical trajectory has been obtained.15 Although initially studied in the outpatient setting, angiotensin converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARB) have been shown in multiple observational studies to be safely and effectively initiated during hospitalization for AHF, with associated reduction in rehospitalization and mortality.16,17

Shifting the Paradigm of Guideline-directed Medical Therapy Initiation

Article image

ARNI is preferred to RAAS-I alone given substantial mortality benefit, similar tolerability, and long-term cost-effectiveness compared to ACEi or ARB.18–20 The PARADIGM-HF trial showed a 16% all-cause death reduction and 21% hazard reduction for HF first hospitalization in the ARNI group compared to the ACEi group.21 Of the patients entering the ARNI ‘run-in’ phase of the trial, less than 5% dropped out as a result of hypotension, renal dysfunction, and hyperkalemia – the three most common reasons for pre-trial drop-out.22 Patients who were then randomized to ARNI actually had lower rates of discontinuation compared to those randomized to ACEi.21 Importantly, symptomatic hypotension is the most common adverse effect of ARNI initiation and occurs about 15% of the time; it is less commonly seen with ACEi or ARB.21,23 A detailed review by Sauer et al. provides further practical guidance on ARNI use.24

The PIONEER-HF trial suggested benefits of ARNI initiation during hospitalization greater than that achieved with ACEi.23 Analysis of the trial showed a 31% lower hazard for cardiovascular (CV) death or rehospitalization events for patients that were initiated on an ARNI during hospitalization versus those initiated on the ACEi, enalapril, in the first 8 weeks.25 PIONEER-HF showed that an ARNI can safely be initiated in patients with a systolic blood pressure (SBP) >100 mmHg for 6 hours, no use of vasodilators or increase in dose of intravenous diuretics in the preceding 6 hours, and no use of inotropes in the preceding 24 hours.23

Contrary to popular belief, RAAS-I/ARNI initiation is well tolerated in cases of mild renal dysfunction, for example, the interquartile range (IQR) of baseline estimated glomerular filtration rate (eGFR) in PIONEER-HF was 47.1–71.5 ml/min/1.73m2.23,26,27 Acute kidney injury (AKI) after starting RAAS-I occurs in ~13% of HF patients but the long-term overall benefit of RAAS inhibition tends to outweigh any reduction in renal function, which is often transient.28 Experts agree that RAAS-I/ARNI should be continued if serum creatinine (SCr) increases <30% after initiation.29 For changes in SCr >30%, RAAS-I/ARNI doses can be reduced or temporarily held to assess for improvement in renal function. Once renal function begins to improve, RAAS-I/ARNI should be re-trialed to assess maximally tolerated dose.

RAAS-I show modest dose responsiveness. High-dose RAAS-I (equivalent to lisinopril 20–40 mg daily) results in a 6% decrease in all-cause mortality compared to low-dose RAAS-I (equivalent to lisinopril 5–10 mg daily).30 This effect is similar for ARNI with maintained advantage over ACEi or ARB even at lower doses.31 Clinicians should aim to initiate a RAAS-I, preferably an ARNI, as soon as clinical stability has been obtained. Table 1 shows our proposed strategy of RAAS-I/ARNI initiation.

β-blockers

In three landmark trials, β-blockers demonstrated a reduction in all-cause mortality by 34–35%, even among patients with severe HF or recent decompensation.32–34 In-hospital initiation of β-blocker therapy for HFrEF has been shown to be safe, well tolerated, and associated with reduced mortality within the first 30 days of hospital discharge.35 This was observed even in patients with signs and symptoms of congestion. β-blockers show the clearest dose response relationship among GDMT.36 Initiating or up-titrating a β-blocker during hospitalization is an effective strategy to increase the likelihood of long-term target dose attainment.37 Martinez-Sellez et al. showed the safety of early initiation of carvedilol in AHF patients at a median 3 days after admission, achieving a mean discharge dose of 23 mg/day among those who tolerated in-hospital up-titration.38 No study has definitively shown the superiority of one evidence-based β-blocker versus another.39 Carvedilol may have a more potent blood pressure lowering effect versus metoprolol succinate in those with more elevated blood pressure.40 Although, for patients with lower blood pressure at baseline, β-blockers are hemodynamically well tolerated.41 In COPERNICUS, patients with a systolic blood pressure (SBP) of 85–95 mmHg at baseline had an increase in SBP with carvedilol greater than placebo.

Optimizing Guideline-directed Medical Therapies for Heart Failure

Article image

β-blockers should not routinely be discontinued on admission for AHF. Multiple studies have shown that discontinuation of β-blockers in AHF is associated with worsening short- and long-term mortality, although hypotension/shock or severe pulmonary edema may represent indications for temporary discontinuation.42 Otherwise, as long as the patient confirms adherence prior to admission, β-blockers can be safely continued during hospitalization.

β-blockers should be initiated or up-titrated so long as the patient is hemodynamically stable without marked volume overload, typically after the initiation of a RAAS-I/ARNI.43 Younger patients with higher BMI and without chronic obstructive pulmonary disease tend to tolerate higher doses and may be appropriate for a higher starting dose or more aggressive up-titration.44 Up-titration in patients may occur as tolerated toward target dose prior to discharge (Table 1).

Pre-discharge Checklist

Article image

Mineralocorticoid Receptor Antagonist

Only one-third of patients indicated for MRA receive them.8 Use of these agents has reduced CV death and HF hospitalization, with benefits observed within 30 days of initiation.45 Addition of MRA to RAAS-I and β-blockers has been shown to be safe during hospitalization.46 Further, long-term adherence is substantially improved with in-hospital initiation compared to delaying therapy to outpatient at clinician discretion.47 Hospitalization represents an opportunity to safely initiate an MRA under close monitoring, with provider fear of hyperkalemia and renal function often cited as barriers to initiation.48 Potassium binding drugs such as patiromer and sodium zirconium cyclosilicate have shown to be effective long-term options to maintain normokalemia in HF patients.49 While these agents do not confer mortality benefit on their own, they are beneficial in patients who would have otherwise been unable to tolerate MRA therapy. Additionally, combined usage of MRA with SGLT2i or ARNI (versus ACEi) lowers the risk of hyperkalemia.50,51 At the dosages commonly used in trials, MRAs do not have any blood pressure lowering effect and can be safely initiated on borderline hypotensive patients.52 There is no clear dose response relationship with MRA, although the target dose in major trials was the equivalent of 25–50 mg/day of spironolactone.53 We suggest initiating an MRA after RAAS-I/ARNI and β-blocker initiation, but prior to discharge (Table 1).

Sodium-Glucose Cotransporter-2 Inhibitor

The DAPA-HF and EMPEROR-Reduced trials showed the significant benefits of SGLT2i in the treatment of patients with HFrEF, including reductions in all-cause mortality risk by 13% and recurrent CV hospitalizations by 25% in HFrEF patients with or without type 2 diabetes (T2DM).6 These clinical benefits can be seen within 12 days of initiation.54 Additionally, the DAPA-CKD trial showed a 44% decrease in the composite of a sustained decline in eGFR of at least 50%, end-stage kidney disease, or death from renal causes.55 Similar results were found in the CREDENCE trial.56 With an estimated 54% of HFrEF patients having concomitant CKD and declining renal function as one of the strongest predictors of mortality in HFrEF, SGLT2i hold substantial clinical promise as a vital fourth pillar of GDMT.57

The novelty of SGLT2i, along with their potential impact on glycemic control for patients with T2DM contribute to relatively low rates of prescription for HFrEF compared to more established pillars of GDMT.58,59 An eGFR <30 ml/min/1.73 m2 is one of a few exclusions to starting the medication at this time, but more than 80% of HFrEF patients are eligible for treatment with SGLT2i.7 SGLT2i can be safely initiated during hospitalization and do not need to be up-titrated from its initial dosage. Sotagliflozin, a dual SGLT-1 and -2 inhibitor, displayed benefits within 1 month of initiation when started during or shortly after hospitalization.60–62

SGLT2i do exhibit a modest BP lowering effect in HF patients (~1–4mmHg).63,64 Additionally, attention should be paid to glycemic management when initiating SGLT-2i in patients on sulfonylureas or insulin due to risk of hypoglycemia and euglycemic diabetic ketoacidosis.64 Volume status of patients on diuretics should be reevaluated due to the risk of over-diuresis. Honigberg et al. previously described in detail the practical considerations for the initiation of SGLT2i. Given the rapid onset of clinical benefit, we favor in-hospital initiation of SGLT2i.65 A summary of the important points for in-hospital initiation are presented in Table 1.

Hydralazine/Nitrate

Although the hydralazine/nitrate combination was the first treatment to show mortality benefit in HFrEF, its usage varies widely.66,67 Current guidelines recommend a hydralazine/nitrate combination for New York Heart Association (NYHA) class III/IV self-described African-American patients who are optimized on RAAS-I and β-blockers.5 Because of modest mortality benefits and relatively larger blood pressure effects, the hydralazine/nitrate combination does not need to be prioritized for in-hospital initiation and is not one of the critical four pillars of therapy.67,68 In patients who cannot take RAAS-I/ARNI due to renal dysfunction, hyperkalemia, or drug intolerance, the hydralazine/nitrate combination can be considered as alternate therapy, regardless of race.5 However, because of the demonstrated superiority of RAAS-I/ARNI, a repeat trial with a RAAS-I/ARNI should be attempted when deemed safe.68

Loop Diuretic

Diuretic optimization is an important aspect of chronic HF treatment for symptom management and reducing the likelihood of hospitalization for worsening HF.69 Three loop diuretics are commonly used for the treatment of HF: furosemide, bumetanide and torsemide.70 No trials have definitively shown superiority of one versus the other; however, furosemide is most commonly used in clinical practice and generally has the lowest acquisition cost.71,72 In a meta-analysis, torsemide has been shown to reduce hospitalizations, lower cardiac mortality, and improve functional status compared to furosemide.73 Torsemide also has the longest half-life and offers the advantage of consistent diuretic effect with once daily dosing.71 Both torsemide and bumetanide have superior bioavailability to furosemide. Guidelines recommend trialing an oral diuretic prior to discharge to verify dose effectiveness.5 In a study comparing patients observed on at least 24 hours of an oral diuretic regimen prior to discharge versus those not, 30-, 60-, and 90-day HF readmissions were reduced significantly.74 Fluid restriction should be liberalized for an oral diuretic trial to better match the diuretic dose with the patient’s unrestricted fluid intake.75 The lowest effective dose of loop diuretic should be used to maintain euvolemia, allowing for maximal up-titration of GDMT.71

Transition of Care

Although the average length of stay for an AHF admission is 5 days, an early focus on GDMT during the admission can increase the likelihood of initiation of all four agents, especially with increasing system pressures to reduce time in hospital.76,77 Following discharge, maintaining momentum and ensuring safety of in-hospital initiation and titration of GDMT is dependent on successful transition of care to the outpatient setting. A systematic review identified eight common components of successful programs: “telephone follow-up, education, self-management, weight monitoring, sodium restriction or dietary advice, exercise recommendations, medication review, and social and psychological support.”78 Supervised exercise training programs (cardiac rehabilitation) for at least 8 weeks following discharge reduce mortality and rehospitalization.79 Iron deficiency should be treated prior to discharge, with one dose of intravenous iron prior to discharge and additional doses as an outpatient shown to improve symptoms and reduce risk of rehospitalization.43,80 Additionally, laboratory values, particularly renal function markers, potassium, and glucose, should be monitored within 1–2 weeks of discharge to evaluate for adverse reactions to GDMT initiation.5 Brain natriuretic peptide (BNP) or N-terminal pro-hormone BNP (NT-proBNP) levels obtained at 1–2 months after GDMT initiation can be useful predictors of rehospitalization and response to therapy.18,81 An assessment of left ventricular systolic function by echocardiography should be performed ~3 months after optimal GDMT has been reached to assess response to therapy and indication for ICD implantation.43 Further, the coronavirus disease 2019 pandemic has highlighted the opportunities to integrate telehealth to continue the aggressive up-titration of GDMT with the use of specialized nurses and pharmacists.82 A suggested pre-discharge checklist is presented in Figure 2 and strategies to overcome common clinical barriers are presented in Table 2.

Overcoming Common Clinical Barriers to Guideline-directed Medical Therapies Optimization

Article image

Conclusion

Given the clinical benefits seen within 30 days of initiation of RAAS-I/ARNI, β-blockers, MRA, and SGLT2i, combined initiation of these four foundational therapies is preferable to the outdated paradigm of pursuing maximally tolerated β-blockers and RAAS-I prior to the addition of secondary agents. Additionally, due to the severe lack of optimization of GDMT in the outpatient setting, in-hospital initiation and titration of the four agents should be attempted in all patients admitted for AHF. The ability to closely monitor in the hospital setting allows for more aggressive up-titration and early recognition of adverse effects. Our strategy prioritizes the implementation of all four pillars of GDMT to meet the evolving standard of optimal treatment of HFrEF.

References

  1. Heidenreich PA, Albert NM, Allen LA, et al. Forecasting the impact of heart failure in the United States: a policy statement from the American Heart Association. Circ Heart Fail 2013;6:606–19.
    Crossref | PubMed
  2. Urbich M, Globe G, Pantiri K, et al. A systematic review of medical costs associated with heart failure in the USA (2014–2020). Pharmacoeconomics 2020;38:1219–36.
    Crossref | PubMed
  3. Blecker S, Paul M, Taksler G, et al. Heart failure-associated hospitalizations in the United States. J Am Coll Cardiol 2013;61:1259–67.
    PubMed
  4. Owan TE, Hodge DO, Herges RM, et al. Trends in prevalence and outcome of heart failure with preserved ejection fraction. N Engl J Med 2006;355:251–9.
    Crossref | PubMed
  5. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2013;62:e147–239.
    Crossref | PubMed
  6. Zannad F, Ferreira JP, Pocock SJ, et al. SGLT2 inhibitors in patients with heart failure with reduced ejection fraction: a meta-analysis of the EMPEROR-Reduced and DAPA-HF trials. Lancet 2020;396:819–29.
    Crossref | PubMed
  7. 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;396:121–8.
    Crossref | PubMed
  8. Greene SJ, Butler J, Albert NM, et al. Medical therapy for heart failure with reduced ejection fraction: the CHAMP-HF Registry. J Am Coll Cardiol 2018;72:351–66.
    Crossref | PubMed
  9. Gracia E, Hamid A, Butler J. Timely management of new-onset heart failure. Circulation 2019;140:621–3.
    Crossref | PubMed
  10. Roth GA, Poole JE, Zaha R, et al. Use of guideline-directed medications for heart failure before cardioverter-defibrillator implantation. J Am Coll Cardiol 2016;67:1062–9.
    Crossref | PubMed
  11. Hancock HC, Close H, Fuat A, et al. Barriers to accurate diagnosis and effective management of heart failure have not changed in the past 10 years: a qualitative study and national survey. BMJ Open 2014;4:e003866.
    Crossref | PubMed
  12. Piamjariyakul U, Yadrich DM, Russell C, et al. Patients’ annual income adequacy, insurance premiums and out-of-pocket expenses related to heart failure care. Heart Lung 2014;43:469–75.
    Crossref | PubMed
  13. Yamaguchi T, Kitai T, Miyamoto T, et al. Effect of optimizing guideline-directed medical therapy before discharge on mortality and heart failure readmission in patients hospitalized with heart failure with reduced ejection fraction. Am J Cardiol 2018;121:969–74.
    Crossref | PubMed
  14. Fonarow GC, Abraham WT, Albert NM, et al. Prospective evaluation of beta-blocker use at the time of hospital discharge as a heart failure performance measure: results from OPTIMIZE-HF. J Card Fail 2007;13:722–31.
    Crossref | PubMed
  15. Hollenberg SM, Stevenson LW, Ahmad T, et al. 2019 ACC expert consensus decision pathway on risk assessment, management, and clinical trajectory of patients hospitalized with heart failure: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2019;74:1966–2011.
    Crossref | PubMed
  16. Gilstrap LG, Fonarow GC, Desai AS, et al. Initiation, continuation, or withdrawal of angiotensin-converting enzyme inhibitors/angiotensin receptor blockers and outcomes in patients hospitalized with heart failure with reduced ejection fraction. J Am Heart Assoc 2017;6:e004675.
    Crossref | PubMed
  17. Sanam K, Bhatia V, Parvataneni S, et al. Discharge initiation of ACE inhibitors or ARBS is associated with significantly lower 30-day all-cause readmission in hospitalized older patients with heart failure and reduced ejection fraction. J Am Coll Cardiol 2014;63(12 Suppl):A562.
    Crossref
  18. DeVore AD, Braunwald E, Morrow DA, et al. Initiation of angiotensin-neprilysin inhibition after acute decompensated heart failure: secondary analysis of the open-label extension of the PIONEER-HF Trial. JAMA Cardiol 2020;5:202–7.
    Crossref | PubMed
  19. Gaziano TA, Fonarow GC, Velazquez EJ, et al. Cost-effectiveness of sacubitril-valsartan in hospitalized patients who have heart failure with reduced ejection fraction. JAMA Cardiol 2020;5:1236–44.
    Crossref | PubMed
  20. Gaziano TA, Fonarow GC, Claggett B, et al. Cost-effectiveness analysis of sacubitril/valsartan vs enalapril in patients with heart failure and reduced ejection fraction. JAMA Cardiol 2016;1:666–72.
    Crossref | PubMed
  21. McMurray JJ, Packer M, Desai AS, et al. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 2014;371:993–1004.
    Crossref | PubMed
  22. Desai AS, Solomon S, Claggett B, et al. Factors associated with noncompletion during the run-in period before randomization and influence on the estimated benefit of LCZ696 in the PARADIGM-HF trial. Circ Heart Fail 2016;9: e002735.
    Crossref | PubMed
  23. Velazquez EJ, Morrow DA, DeVore AD, et al. Angiotensin-neprilysin inhibition in acute decompensated heart failure. N Engl J Med 2019;380:539–48.
    Crossref | PubMed
  24. Sauer AJ, Cole R, Jensen BC, et al. Practical guidance on the use of sacubitril/valsartan for heart failure. Heart Fail Rev 2019;24:167–76.
    Crossref | PubMed
  25. DeVore AD, Braunwald E, Morrow DA, et al. Initiation of angiotensin-neprilysin inhibition after acute decompensated heart failure: secondary analysis of the open-label extension of the PIONEER-HF trial. JAMA Cardiol 2020;5:202–7.
    Crossref | PubMed
  26. Akerman CC, Beavers JC. Risk factors for intolerance of inpatient sacubitril/valsartan initiation. J Pharm Pract 2019; epub ahead of press.
    Crossref | PubMed
  27. Kozhuharov N, Goudev A, Flores D, et al. Effect of a strategy of comprehensive vasodilation vs usual care on mortality and heart failure rehospitalization among patients with acute heart failure: the GALACTIC randomized clinical Trial. JAMA 2019;322:2292–302.
    Crossref | PubMed
  28. Beldhuis IE, Streng KW, Ter Maaten JM, et al. Renin-angiotensin system inhibition, worsening renal function, and outcome in heart failure patients with reduced and preserved ejection fraction: a meta-analysis of published study data. Circ Heart Fail 2017;10.
    Crossref | PubMed
  29. Clark AL, Kalra PR, Petrie MC, et al. Change in renal function associated with drug treatment in heart failure: national guidance. Heart 2019;105:904–10.
    Crossref | PubMed
  30. Khan MS, Fonarow GC, Ahmed A, et al. Dose of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers and outcomes in heart failure: a meta-analysis. Circ Heart Fail 2017;10.
    Crossref | PubMed
  31. Vardeny O, Claggett B, Packer M, et al. Efficacy of sacubitril/valsartan vs. enalapril at lower than target doses in heart failure with reduced ejection fraction: the PARADIGM-HF trial. Eur J Heart Fail 2016;18:1228–34.
    Crossref | PubMed
  32. Packer M, Fowler MB, Roecker EB, et al. Effect of carvedilol on the morbidity of patients with severe chronic heart failure: results of the carvedilol prospective randomized cumulative survival (COPERNICUS) study. Circulation 2002;106:2194–9.
    Crossref | PubMed
  33. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet 1999;353:9–13.
    Crossref | PubMed
  34. Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet 1999;353:2001–7.
    Crossref | PubMed
  35. Bhatia V, Bajaj NS, Sanam K, et al. Beta-blocker use and 30-day all-cause readmission in medicare beneficiaries with systolic heart failure. Am J Med 2015;128:715–21.
    Crossref | PubMed
  36. Fiuzat M, Wojdyla D, Kitzman D, et al. Relationship of beta-blocker dose with outcomes in ambulatory heart failure patients with systolic dysfunction: results from the HF-ACTION (Heart Failure: A Controlled Trial Investigating Outcomes of Exercise Training) trial. J Am Coll Cardiol 2012;60:208–15.
    Crossref | PubMed
  37. Gattis WA, O’Connor CM, Gallup DS, et al. Predischarge initiation of carvedilol in patients hospitalized for decompensated heart failure: results of the Initiation Management Predischarge: Process for Assessment of Carvedilol Therapy in Heart Failure (IMPACT-HF) trial. J Am Coll Cardiol 2004;43:1534–41.
    Crossref | PubMed
  38. Martinez-Selles M, Datino T, Alhama M, et al. Rapid carvedilol up-titration in hospitalized patients with systolic heart failure. J Heart Lung Transplant 2008;27:914–16.
    Crossref | PubMed
  39. Fröhlich H, Zhao J, Täger T, et al. Carvedilol compared with metoprolol succinate in the treatment and prognosis of patients with stable chronic heart failure: Carvedilol or Metoprolol Evaluation Study. Circ Heart Fail 2015;8:887–96.
    Crossref | PubMed
  40. Franz IW, Agrawal B, Wiewel D, Ketelhut R. Comparison of the antihypertensive effects of carvedilol and metoprolol on resting and exercise blood pressure. Clin Investig 1992;70(Suppl 1):S53–7.
    Crossref | PubMed
  41. Rouleau JL, Roecker EB, Tendera M, et al. Influence of pretreatment systolic blood pressure on the effect of carvedilol in patients with severe chronic heart failure: the Carvedilol Prospective Randomized Cumulative Survival (COPERNICUS) study. J Am Coll Cardiol 2004;43:1423–9.
    Crossref | PubMed
  42. Prins KW, Neill JM, Tyler JO, et al. Effects of beta-blocker withdrawal in acute decompensated heart failure: a systematic review and meta-analysis. JACC Heart Failure 2015;3:647–53.
    Crossref | PubMed
  43. Yancy CW, Januzzi JL Jr., Allen LA, et al. 2017 ACC Expert Consensus Decision Pathway for Optimization of Heart Failure Treatment: answers to 10 pivotal issues about heart failure with reduced ejection fraction: a report of the American College of Cardiology Task Force on Expert Consensus Decision Pathways. J Am Coll Cardiol 2018;71:201–30.
    Crossref | PubMed
  44. Cohen-Solal A, Jacobson AF, Piña IL. Beta blocker dose and markers of sympathetic activation in heart failure patients: interrelationships and prognostic significance. ESC Heart Fail 2017;4:499–506.
    Crossref | PubMed
  45. Rossi R, Crupi N, Coppi F, et al. Importance of the time of initiation of mineralocorticoid receptor antagonists on risk of mortality in patients with heart failure. J Renin Angiotensin Aldosterone Syst 2015;16:119–25.
    Crossref | PubMed
  46. Durstenfeld MS, Katz SD, Park H, Blecker S. Mineralocorticoid receptor antagonist use after hospitalization of patients with heart failure and post-discharge outcomes: a single-center retrospective cohort study. BMC Cardiovasc Disord 2019;19:194.
    Crossref | PubMed
  47. Wirtz HS, Sheer R, Honarpour N, et al. Real-world analysis of guideline-based therapy after hospitalization for heart failure. J Am Heart Assoc 2020;9:e015042.
    Crossref | PubMed
  48. Ferreira JP, Rossignol P, Machu JL, et al. Mineralocorticoid receptor antagonist pattern of use in heart failure with reduced ejection fraction: findings from BIOSTAT-CHF. Eur J Heart Fail 2017;19:1284–93.
    Crossref | PubMed
  49. Sidhu K, Sanjanwala R, Zieroth S. Hyperkalemia in heart failure. Curr Opin Cardiol 2020;35:150–5.
    Crossref | PubMed
  50. Desai AS, Vardeny O, Claggett B, et al. Reduced risk of hyperkalemia during treatment of heart failure with mineralocorticoid receptor antagonists by use of sacubitril/valsartan compared with enalapril: a secondary analysis of the PARADIGM-HF Trial. JAMA Cardiol 2017;2:79–85.
    Crossref | PubMed
  51. Kristensen SL, Docherty KF, Jhund PS, et al. Dapagliflozin reduces the risk of hyperkalaemia in patients with heart failure and reduced ejection fraction: a secondary analysis DAPA-HF. Eur Heart J 2020;41(Suppl 2):ehaa946.0939.
    Crossref
  52. Bazoukis G, Thomopoulos C, Tse G, Tsioufis C. Is there a blood pressure lowering effect of MRAs in heart failure? An overview and meta-analysis. Heart Fail Rev 2018;23:547–53.
    Crossref | PubMed
  53. Berbenetz NM, Mrkobrada M. Mineralocorticoid receptor antagonists for heart failure: systematic review and meta-analysis. BMC Cardiovasc Disord 2016;16:246.
    Crossref | PubMed
  54. Packer M, Anker SD, Butler J, et al. Effect of Empagliflozin on the Clinical Stability of Patients with Heart Failure and a Reduced Ejection Fraction: the EMPEROR-Reduced Trial. Circulation 2021;143:326–336.
    Crossref | PubMed
  55. Wheeler DC, Stefansson BV, Batiushin M, et al. The Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease (DAPA-CKD) trial: baseline characteristics. Nephrol Dial Transplant 2020;35:1700–11.
    Crossref | PubMed
  56. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 2019;380:2295–306.
    Crossref | PubMed
  57. McAlister FA, Ezekowitz J, Tarantini L, et al. Renal dysfunction in patients with heart failure with preserved versus reduced ejection fraction: impact of the new Chronic Kidney Disease-Epidemiology Collaboration Group formula. Circ Heart Fail 2012;5:309–14.
    Crossref | PubMed
  58. Vaduganathan M, Sathiyakumar V, Singh A, et al. Prescriber patterns of SGLT2i after expansions of U.S. Food and Drug Administration labeling. J Am Coll Cardiol 2018;72:3370–2.
    Crossref | PubMed
  59. Gao Y, Peterson E, Pagidipati N. Barriers to prescribing glucose-lowering therapies with cardiometabolic benefits. Am Heart J 2020;224:47–53.
    Crossref | PubMed
  60. Bhatt DL, Szarek M, Steg PG, et al. Sotagliflozin in patients with diabetes and recent worsening heart failure. N Engl J Med. 2021;384:117–28.
    Crossref | PubMed
  61. Bhatt DL, Szarek M, Pitt B, et al. Sotagliflozin in patients with diabetes and chronic kidney disease. N Engl J Med 2021;384:129–39.
    Crossref | PubMed
  62. Damman K, Beusekamp JC, Boorsma EM, et al. Randomized, double-blind, placebo-controlled, multicentre pilot study on the effects of empagliflozin on clinical outcomes in patients with acute decompensated heart failure (EMPA-RESPONSE-AHF). Eur J Heart Fail 2020;22:713–22.
    Crossref | PubMed
  63. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 2019;381:1995–2008.
    Crossref | PubMed
  64. Honigberg MC, Vardeny O, Vaduganathan M. Practical considerations for the use of sodium-glucose co-transporter 2 inhibitors in heart failure. Circ Heart Fail 2020;13:e006623.
    Crossref | PubMed
  65. Berg DD, Jhund PS, Docherty KF, et al. Time to clinical benefit of dapagliflozin and significance of prior heart failure hospitalization in patients with heart failure with reduced ejection fraction. JAMA Cardiol 2021.
    Crossref | PubMed
  66. Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration Cooperative Study. N Engl J Med 1986;314:1547–52.
    Crossref | PubMed
  67. Al-Mohammad A. Hydralazine and nitrates in the treatment of heart failure with reduced ejection fraction. ESC Heart Fail 2019;6:878–83.
    Crossref | PubMed
  68. Farag M, Mabote T, Shoaib A, et al. Hydralazine and nitrates alone or combined for the management of chronic heart failure: a systematic review. Int J Cardiol 2015;196:61–9.
    Crossref | PubMed
  69. Faris R, Flather MD, Purcell H, et al. Diuretics for heart failure. Cochrane Database Syst Rev 2006;(1):CD003838.
    Crossref | PubMed
  70. Buggey J, Mentz RJ, Pitt B, et al. A reappraisal of loop diuretic choice in heart failure patients. Am Heart J 2015;169:323–33.
    Crossref | PubMed
  71. Felker GM, Ellison DH, Mullens W, et al. J Am Coll Cardiol 2020;75:1178–95.
    Crossref | PubMed
  72. Mentz RJ, Hasselblad V, DeVore AD, et al. Torsemide versus furosemide in patients with acute heart failure (from the ASCEND-HF trial). Am J Cardiol 2016;117:404–11.
    Crossref | PubMed
  73. Abraham B, Megaly M, Sous M, et al. Meta-analysis comparing torsemide versus furosemide in patients with heart failure. Am J Cardiol 2020;125:92–9.
    Crossref | PubMed
  74. Laliberte B, Reed BN, Devabhakthuni S, et al. Observation of patients transitioned to an oral loop diuretic before discharge and risk of readmission for acute decompensated heart failure. J Card Fail Oct 2017;23:746–52.
    Crossref | PubMed
  75. Li Y, Fu B, Qian X. Liberal versus restricted fluid administration in heart failure patients. A systematic review and meta-analysis of randomized trials. Int Heart J 2015;56:192–5.
    Crossref | PubMed
  76. Sud M, Yu B, Wijeysundera HC, et al. Associations between short or long length of stay and 30-day readmission and mortality in hospitalized patients with heart failure. JACC Heart Fail 2017;5:578–88.
    Crossref | PubMed
  77. Samsky MD, Ambrosy AP, Youngson E, et al. Trends in readmissions and length of stay for patients hospitalized with heart failure in Canada and the United States. JAMA Cardiol 2019;4:444–53.
    Crossref | PubMed
  78. Takeda A, Taylor SJ, Taylor RS, et al. Clinical service organisation for heart failure. Cochrane Database Syst Rev 2012;(9):CD002752.
    Crossref | PubMed
  79. Piepoli MF, Davos C, Francis DP, Coats AJ. Exercise Training Meta-analysis of Trials in Patients with Chronic Heart Failure (ExTraMATCH). BMJ 2004;328:189.
    Crossref | PubMed
  80. 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;396:1895–904.
    Crossref | PubMed
  81. Omar HR, Guglin M. Post-discharge rise in BNP and rehospitalization for heart failure. Herz 2019;44:450–4.
    Crossref | PubMed
  82. Thibodeau JT, Gorodeski EZ. Telehealth for uptitration of guideline-directed medical therapy in heart failure. Circulation 2020;142:1507–9.
    Crossref | PubMed