Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 555
2.
JACC Heart Fail ; 8(5): 359-368, 2020 05.
Article En | MEDLINE | ID: mdl-32171760

OBJECTIVES: This study investigated the effects of a mid-trial protocol amendment requiring elevated natriuretic peptides for inclusion in the COMMANDER-HF (A Study to Assess the Effectiveness and Safety of Rivaroxaban in Reducing the Risk of Death, Myocardial Infarction, or Stroke in Participants with Heart Failure and Coronary Artery Disease Following an Episode of Decompensated Heart Failure) trial. BACKGROUND: Heart failure (HF) trials that select patients based on history of HF hospitalization alone are susceptible to regional variations in event rates. Elevated plasma concentrations of natriuretic peptides (NPs) as selection criteria may help HF ascertainment and risk enrichment. In the COMMANDER-HF trial, B-type natriuretic peptide ≥200 ng/l or N-terminal pro-B-type natriuretic peptide ≥800 ng/l were added to inclusion criteria as a mid-trial protocol amendment, providing a unique case-study of NP-based inclusion criteria. METHODS: We compared the baseline characteristics, event rates, and treatment effects for patients enrolled before and after the NP protocol amendment. The primary endpoint was all-cause death, myocardial infarction, or stroke. Secondary endpoints included HF rehospitalization and cardiovascular death. RESULTS: A total of 5,022 patients with left ventricular ejection fraction ≤40% and coronary artery disease were included. Compared to patients enrolled before the NP protocol amendment, those enrolled post-amendment (n = 3,867, 77%) were older, more often had diabetes, and had lower values for body mass index, left ventricular ejection fraction, and estimated glomerular filtration rate, higher heart rate, and higher event rates: primary endpoint (hazard ratio [HR]: 1.32; 95% confidence interval [CI]: 1.16 to 1.50), cardiovascular death (HR: 1.29; 95% CI: 1.11 to 1.50), HF rehospitalization (HR: 1.31; 95% CI: 1.15 to 1.49), and major bleeding (HR: 1.71; 95% CI: 1.11 to 2.65). Differences between pre- and post-amendment rates were confined to and driven by Eastern Europe. This protocol amendment did not modify the neutral effect of rivaroxaban on the primary endpoint (p interaction = 0.36) or secondary endpoints. CONCLUSIONS: In a global event-driven trial of rivaroxaban in HF, requiring elevated NPs for inclusion increased event rates allowing earlier completion of the trial but did not modify treatment effect. These data inform future HF trials regarding the expected impact of NP-based inclusion criteria on patient characteristics and event rates. (COMMANDER HF [A Study to Assess the Effectiveness and Safety of Rivaroxaban in Reducing the Risk of Death, Myocardial Infarction, or Stroke in Participants With Heart Failure and Coronary Artery Disease Following an Episode of Decompensated Heart Failure] NCT01877915).


Heart Failure/drug therapy , Natriuretic Peptides/blood , Patient Selection , Rivaroxaban/therapeutic use , Stroke Volume/physiology , Ventricular Function, Left/physiology , Aged , Biomarkers/blood , Double-Blind Method , Factor Xa Inhibitors/therapeutic use , Female , Global Health , Heart Failure/blood , Heart Failure/mortality , Humans , Male , Middle Aged , Prognosis , Survival Rate/trends
3.
J Card Fail ; 26(5): 429-437, 2020 May.
Article En | MEDLINE | ID: mdl-32068002

BACKGROUND: Elamipretide, a novel mitochondrial modulating agent, improves myocardial energetics; however, it is unknown whether this mechanistic benefit translates into improved cardiac structure and function in heart failure (HF) with reduced ejection fraction (HFrEF). The objective of this study was to evaluate the effects of multiple subcutaneous doses of elamipretide on left ventricular end systolic volume (LVESV) as assessed by cardiac magnetic resonance imaging. METHODS: We randomized 71 patients with HFrEF (LVEF ≤ 40%) in a double-blind, placebo-controlled trial in a 1:1:1 ratio to receive placebo, 4 mg or 40 mg elamipretide once daily for 28 consecutive days. RESULTS: The mean age (standard deviation) of the study population was 65 ± 10 years, 24% were females, and the mean EF was 31% ± 7%. The change in LVESV from baseline to week 4 was not significantly different between elamipretide 4 mg (89.4 mL to 85 mL; difference, -4.4 mL) or 40 mg (77.9 mL to 76.6 mL; difference, -1.2 mL) compared with placebo (77.7 mL to 74.6 mL; difference, -3.8 mL) (4 mg vs placebo: difference of means, -0.3; 95% CI, -4.6 to 4.0; P = 0.90; and 40 mg vs placebo: difference of means, 2.3; 95% CI, -1.9 to 6.5; P  =  0.28). Also, no significant differences in change in LVESV and LVEF were observed between placebo and either of the elamipretide groups. Rates of any study drug-related adverse events were similar in the 3 groups. CONCLUSIONS: Elamipretide was well tolerated but did not improve LVESV at 4 weeks in patients with stable HFrEF compared with placebo.


Heart Failure , Ventricular Function, Left , Aged , Female , Heart Failure/drug therapy , Humans , Middle Aged , Oligopeptides , Stroke Volume
4.
Clin J Am Soc Nephrol ; 14(11): 1670-1676, 2019 11 07.
Article En | MEDLINE | ID: mdl-31554619

The pretransplant risk assessment for patients with ESKD who are undergoing evaluation for kidney transplant is complex and multifaceted. When considering cardiovascular disease in particular, many factors should be considered. Given the increasing incidence of kidney transplantation and the growing body of evidence addressing ESKD-specific cardiovascular risk profiles, there is an important need for a consolidated, evidence-based model that considers the unique cardiovascular challenges that these patients face. Cardiovascular physiology is altered in these patients by abrupt shifts in volume status, altered calcium-phosphate metabolism, high-output states (in the setting of arteriovenous fistulization), and adverse geometric and electrical remodeling, to name a few. Here, we present a contemporary review by addressing cardiomyopathy/heart failure, pulmonary hypertension, valvular dysfunction, and arrhythmia/sudden cardiac death within the ESKD population.


Cardiovascular Diseases/complications , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/surgery , Kidney Transplantation , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/therapy , Humans , Practice Guidelines as Topic , Preoperative Period , Risk Assessment
5.
Clin Pharmacol Drug Dev ; 8(7): 942-951, 2019 10.
Article En | MEDLINE | ID: mdl-30452784

The chymase inhibitor fulacimstat is developed as a first-in-class treatment option for the inhibition of adverse cardiac remodeling in patients with left ventricular dysfunction (LVD) after acute myocardial infarction (MI). The aim of the study was to examine the safety and tolerability of fulacimstat in patients with LVD after remote MI. A multicenter, multinational randomized, placebo-controlled study was performed in clinically stable patients (40-79 years of age, left ventricular ejection fraction ≤ 45% because of MI in medical history) who were on stable evidence-based standard-of-care therapies for LVD post-MI including an angiotensin converting enzyme inhibitor or angiotensin receptor blocker at doses of at least half the recommended target dose. Patients were treated for 2 weeks with either placebo (n = 12) or 4 different doses of fulacimstat (5 mg twice daily, n = 9; 10 mg twice daily, n = 9; 25 mg twice daily, n = 10; 50 mg once daily, n = 9). Fulacimstat was safe and well tolerated at all examined doses. There were no clinically relevant effects on vital signs or potassium levels compared with placebo treatment. Mean plasma concentrations of fulacimstat increased with the administered dose and reached exposures predicted to be therapeutically active. The safety profile and the absence of effects on blood pressure or heart rate in a chronic patient population having similar comorbidities and receiving similar comedication as patients after acute MI support future clinical trials with fulacimstat in patients after acute MI.


Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Carboxylic Acids/administration & dosage , Heart Failure/prevention & control , Indenes/administration & dosage , Myocardial Infarction/complications , Pyrimidines/administration & dosage , Ventricular Dysfunction, Left/drug therapy , Adult , Aged , Carboxylic Acids/adverse effects , Carboxylic Acids/pharmacokinetics , Chymases/antagonists & inhibitors , Drug Administration Schedule , Female , Heart Failure/etiology , Humans , Indenes/adverse effects , Indenes/pharmacokinetics , Male , Middle Aged , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Treatment Outcome , Ventricular Dysfunction, Left/etiology
6.
Eur J Heart Fail ; 21(3): 286-296, 2019 03.
Article En | MEDLINE | ID: mdl-30537163

Multiple drug classes have shown incremental benefits in heart failure with reduced ejection fraction. Most of these trials were designed to achieve specific doses of the investigational agent. Clinical practice guidelines recommend using the same target dosing of therapies, as tolerated. However, with the increasing number of available therapies, clinicians face the challenge of simultaneously using several drugs, achieving target doses, and managing side effects that are often overlapping. Blood pressure, renal function, hyperkalaemia, and other factors may impede achieving target doses of all medications, leaving clinicians with dilemmas as to how to sequence and dose these various classes of drugs. The guideline-directed eligibility for certain drugs and devices requires stability on maximally tolerated doses of background therapies. However, significant variability exists in dosing achieved in clinical practice. We discuss the existing background data regarding the doses of heart failure medications in clinical trials and in practice, and provide recommendations on how to navigate this complex therapeutic decision-making.


Cardiovascular Agents/pharmacology , Heart Failure , Dose-Response Relationship, Drug , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Patient Selection , Practice Guidelines as Topic/standards , Stroke Volume
7.
J Cardiovasc Med (Hagerstown) ; 20(2): 51-58, 2019 Feb.
Article En | MEDLINE | ID: mdl-30540647

: Patients with end-stage renal disease (ESRD) undergoing evaluation for kidney and/or pancreas transplantation represent a population with unique cardiovascular (CV) profiles and unique therapeutic needs. Coronary artery disease (CAD) is common in patients with ESRD, mediated by both the overrepresentation and higher prognostic value of traditional CV risk factors amongst this population, as well as altered cardiovascular responses to failing renal function, likely mediated by dysregulation of the renin-angiotensin-aldosterone system (RAAS) and abnormal calcium and phosphate metabolism. Within the ESRD population, obstructive CAD correlates highly with adverse coronary events, including during the peri-transplant period, and successful revascularization may attenuate some of that increased risk. Accordingly, peri-transplant coronary risk assessment is critical to ensuring optimal outcomes for these patients. The following provides a review of CAD in patients being evaluated for kidney and/or pancreas transplantation, as well as evidence-based recommendations for appropriate peri-transplant evaluation and management.


Coronary Artery Disease/therapy , Kidney Failure, Chronic/surgery , Kidney Transplantation , Pancreas Transplantation , Pancreatic Diseases/surgery , Algorithms , Clinical Decision-Making , Coronary Artery Disease/complications , Coronary Artery Disease/diagnosis , Coronary Artery Disease/physiopathology , Decision Support Techniques , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/physiopathology , Kidney Transplantation/adverse effects , Pancreas Transplantation/adverse effects , Pancreatic Diseases/complications , Pancreatic Diseases/diagnosis , Pancreatic Diseases/physiopathology , Risk Assessment , Risk Factors , Treatment Outcome
8.
Eur J Heart Fail ; 20(11): 1601-1610, 2018 11.
Article En | MEDLINE | ID: mdl-30225882

Despite major advances in the treatment of chronic heart failure (HF) with reduced ejection fraction (HFrEF), morbidity and mortality associated with the condition remain high, suggesting the need for additional treatment options, particularly haemodynamically neutral treatments that do not alter blood pressure, heart rate, or renal function. HF with preserved ejection fraction (HFpEF) is also associated with high morbidity and mortality and adequate treatment options are limited; thus there is a critical unmet need for the development of novel therapies for HFpEF. Chronic HFrEF and HFpEF are both systemic disorders that affect not only the heart but several other tissues and organs including skeletal muscle, leading to exercise intolerance and dyspnoea. Partial adenosine A1-receptor agonists represent a novel potential therapy for HF regardless of underlying ejection fraction given their minimal effect on heart rate and blood pressure, and preclinical data demonstrate several possible beneficial mechanisms, including improved mitochondrial function and sarcoplasmic reticulum Ca2+ -ATPase (SERCA2a) activity, enhanced energy substrate utilization, reverse ventricular remodelling, and anti-ischemic, cardioprotective properties. However, data on this class of drugs in humans are scarce, and the optimal dose of the partial adenosine A1 receptor, neladenoson bialanate, has not been defined. Here we describe the design and rationale of two randomized, double-blind, placebo-controlled, parallel-group, dose-finding phase 2b trials, PANTHEON (HFrEF) and PANACHE (HFpEF), that will advance our understanding of the potential benefit and optimal dose of neladenoson bialanate and provide critical information for the planning of future phase 3 trials.


Adenosine A1 Receptor Agonists/administration & dosage , Dipeptides/administration & dosage , Heart Failure/drug therapy , Pyridines/administration & dosage , Stroke Volume/physiology , Ventricular Function, Left/physiology , Adolescent , Adult , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follow-Up Studies , Heart Failure/physiopathology , Heart Rate/drug effects , Heart Rate/physiology , Humans , Male , Middle Aged , Stroke Volume/drug effects , Treatment Outcome , Young Adult
9.
J Card Fail ; 24(11): 783-792, 2018 11.
Article En | MEDLINE | ID: mdl-30217774

The prevalence of heart failure continues to grow, and this is accompanied by an increase in hospitalization for acute heart failure. Hospitalization for heart failure results in a trajectory shift of the syndrome and is associated with worsening outcomes, increased mortality risk, and high costs. Numerous clinical trials over the past 2 decades have had limited success, with no single agent shown to improve mortality risk. The lack of success is multifactorial and in part related to inadequate targets and end points selected for intervention, underscoring the need to better understand and define the pathophysiology of acute heart failure. To better inform future drug development, this review critically explores the short-term end points and outcomes that previous phase III acute heart failure trials have examined.


Clinical Trials as Topic , Endpoint Determination/methods , Heart Failure/epidemiology , Hospitalization/statistics & numerical data , Acute Disease , Global Health , Heart Failure/therapy , Humans , Prevalence , Survival Rate/trends
10.
N Engl J Med ; 379(14): 1332-1342, 2018 10 04.
Article En | MEDLINE | ID: mdl-30146935

BACKGROUND: Heart failure is associated with activation of thrombin-related pathways, which predicts a poor prognosis. We hypothesized that treatment with rivaroxaban, a factor Xa inhibitor, could reduce thrombin generation and improve outcomes for patients with worsening chronic heart failure and underlying coronary artery disease. METHODS: In this double-blind, randomized trial, 5022 patients who had chronic heart failure, a left ventricular ejection fraction of 40% or less, coronary artery disease, and elevated plasma concentrations of natriuretic peptides and who did not have atrial fibrillation were randomly assigned to receive rivaroxaban at a dose of 2.5 mg twice daily or placebo in addition to standard care after treatment for an episode of worsening heart failure. The primary efficacy outcome was the composite of death from any cause, myocardial infarction, or stroke. The principal safety outcome was fatal bleeding or bleeding into a critical space with a potential for causing permanent disability. RESULTS: Over a median follow-up period of 21.1 months, the primary end point occurred in 626 (25.0%) of 2507 patients assigned to rivaroxaban and in 658 (26.2%) of 2515 patients assigned to placebo (hazard ratio, 0.94; 95% confidence interval [CI], 0.84 to 1.05; P=0.27). No significant difference in all-cause mortality was noted between the rivaroxaban group and the placebo group (21.8% and 22.1%, respectively; hazard ratio, 0.98; 95% CI, 0.87 to 1.10). The principal safety outcome occurred in 18 patients who took rivaroxaban and in 23 who took placebo (hazard ratio, 0.80; 95% CI, 0.43 to 1.49; P=0.48). CONCLUSIONS: Rivaroxaban at a dose of 2.5 mg twice daily was not associated with a significantly lower rate of death, myocardial infarction, or stroke than placebo among patients with worsening chronic heart failure, reduced left ventricular ejection fraction, coronary artery disease, and no atrial fibrillation. (Funded by Janssen Research and Development; COMMANDER HF ClinicalTrials.gov number, NCT01877915 .).


Coronary Artery Disease/drug therapy , Factor Xa Inhibitors/therapeutic use , Heart Failure/drug therapy , Rivaroxaban/therapeutic use , Aged , Coronary Artery Disease/complications , Double-Blind Method , Drug Therapy, Combination , Factor Xa Inhibitors/adverse effects , Female , Follow-Up Studies , Heart Failure/complications , Heart Failure/mortality , Heart Failure/physiopathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Myocardial Infarction/epidemiology , Myocardial Infarction/prevention & control , Natriuretic Peptide, Brain/blood , Patient Readmission/statistics & numerical data , Platelet Aggregation Inhibitors/therapeutic use , Rivaroxaban/adverse effects , Stroke/epidemiology , Stroke/prevention & control , Stroke Volume , Treatment Failure
11.
Int J Cardiol ; 269: 265-271, 2018 Oct 15.
Article En | MEDLINE | ID: mdl-29921516

Cardiovascular disease is the leading cause of mortality and morbidity globally. With widespread and growing use of smart phones and mobile devices, the use of mobile health (mHealth) in transmission of physiologic parameters and patient-referred symptoms to healthcare providers and researchers, as well as reminders and care plan applications from providers to patients, has potential to revolutionize both clinical care and the conduct of clinical trials with improved designs, data capture, and potentially lower costs. In randomized early phase proof-of-concept studies, focusing on lifestyle intervention, there is evidence that mHealth technology can improve outcomes. By contrast, results from small randomized controlled trials that tested mHealth interventions in heart failure patients were disappointing with inconsistent findings. These inconclusive results could be partially attributed to a lack of methodological rigor (insufficient sample size, quasi-experimental design, inadequate mHealth equipment). Therefore, there is an urgent need to develop systematic evidence-based guidelines and parameters for mHealth to be effectively utilized in cardiovascular clinical trials.


Biomedical Research/trends , Cardiovascular Diseases/therapy , Telemedicine/trends , Text Messaging/trends , Biomedical Research/methods , Cardiovascular Diseases/diagnosis , Humans , Observational Studies as Topic/methods , Randomized Controlled Trials as Topic/methods , Telemedicine/methods
12.
J Cardiovasc Med (Hagerstown) ; 19(8): 411-421, 2018 Aug.
Article En | MEDLINE | ID: mdl-29952846

: Despite recent advances in chronic heart failure treatment, prognosis of acute heart failure patients remains poor with a heart failure rehospitalization rate or death reaching approximately 25% during the first 6 months after discharge. In addition, about half of these patients have preserved ejection fraction for which there are no evidence-based therapies. Disappointing results from heart failure clinical trials over the past 20 years emphasize the need for developing novel approaches and pathways for testing new heart failure drugs and devices. Indeed, many trials are being conducted without matching the mechanism and action of the drug with the clinical event. The implementation of these novel approaches should be coupled with the training of a new generation of heart failure physicians and scientists in the art and science of clinical trials. Currently, drug development is led by opinion leaders and experts who, despite their huge personal experience, were never trained systematically on drug development. The aim of this article is to propose a training program of 'drug development in Heart Failure'. A physician attending this course would have to be trained with a major emphasis on heart failure pathophysiology to better match mechanisms of death and rehospitalization with mechanism of action of the drug. Applicants will have to prove their qualifications and special interest in heart failure drug development before enrollment. This article should serve as a roadmap on how to apply emerging general principles in an innovative drug-development-in-heart-failure-process as well as the introduction of a new educational and mentorship program focusing on younger generations of researchers.


Biomedical Research/education , Cardiologists/education , Drug Development/education , Heart Failure/drug therapy , Chronic Disease , Clinical Trials as Topic , Health Knowledge, Attitudes, Practice , Heart Failure/physiopathology , Humans , Mentors , Patient Readmission/statistics & numerical data
13.
Am J Cardiol ; 122(2): 255-260, 2018 07 15.
Article En | MEDLINE | ID: mdl-29731121

Patients with chronic heart failure with reduced ejection fraction (HFrEF) benefit from medical and device therapies targeting sudden cardiac death (SCD). Contemporary estimates of SCD risk after hospitalization for heart failure are limited. We describe the incidence, timing, and clinical predictors of SCD after hospitalization for HFrEF (≤40%) in the EVEREST (Efficacy of Vasopressin Antagonism in Heart Failure Outcome Study with Tolvaptan) trial. Multiple logistic regression analyses tested >30 baseline covariates (including treatment randomization, demographics, comorbid conditions, natriuretic peptides, ejection fraction, and medical and device therapies) to identify predictors of 1-year SCD. Of the 4,024 trial patients discharged alive (97%), there were 268 who experienced SCD (7%) and 703 who experienced non-SCD (17%) during median follow-up of 9.9 months. Implantable cardioverter defibrillator use at baseline was 14.5%. Estimates of SCD at 1, 3, 6, and 12 months were 0.8%, 2.3%, 4.1%, and 7.4%, respectively. Most patients were readmitted before SCD (n = 147, 55%). Male gender, black race, diabetes mellitus, and angiotensin-converting enzyme inhibitor/angiotensin II receptor blocker use were potential predictors of 1-year SCD after hospitalization for HFrEF (all p <0.10); however, this final model demonstrated poor discrimination (C-statistic 0.57). In conclusion, in the EVEREST trial, patients hospitalized for HFrEF faced risks of 1-year postdischarge SCD of 7%, which accrued gradually over time, and were balanced with high competing risks of nonsudden death (17%). Traditional clinical characteristics fail to adequately predict SCD risk. Further data are needed to identify patients at greatest relative risk for SCD (compared with non-SCD) after hospitalization for HFrEF.


Death, Sudden, Cardiac/epidemiology , Defibrillators, Implantable , Heart Failure/mortality , Hospitalization/statistics & numerical data , Stroke Volume/physiology , Tolvaptan/therapeutic use , Aged , Antidiuretic Hormone Receptor Antagonists/therapeutic use , Double-Blind Method , Female , Follow-Up Studies , Heart Failure/physiopathology , Heart Failure/therapy , Hospital Mortality/trends , Humans , Incidence , Male , Risk Factors , Survival Rate/trends , United States/epidemiology
14.
Heart Fail Rev ; 23(4): 597-607, 2018 07.
Article En | MEDLINE | ID: mdl-29611010

Heart failure is a complex disease process, the manifestation of various cardiac and noncardiac abnormalities. General treatment approaches for heart failure have remained the same over the past decades despite the advent of novel therapies and monitoring modalities. In the same vein, the readmission rates for heart failure patients remain high and portend a poor prognosis for morbidity and mortality. In this context, development and implementation of improved algorithms for assessing and treating HF patients during hospitalization remains an unmet need. We propose an expanded algorithm for both monitoring and treating patients admitted for acute decompensated heart failure with the goal to improve post-discharge outcomes and decrease rates of rehospitalizations.


Algorithms , Disease Management , Heart Failure/therapy , Stroke Volume/physiology , Acute Disease , Heart Failure/physiopathology , Hospitalization , Humans
15.
J Card Fail ; 24(2): 112-114, 2018 02.
Article En | MEDLINE | ID: mdl-29329950

Stimulation of the V1a receptor for arginine vasopressin produces myocardial and vascular effects similar to those of angiotensin II while stimulation of the V2 receptor causes fluid retention. There are no data with sustained blockade of the V1a receptor while single-dose experiments suggest benefit. Acute and chronic administration of selective V2 receptor antagonists reliably relieves dyspnea and produces diuresis without adverse effects on renal function or neurohormonal stimulation, either as adjunctive or alternative therapy to loop diuretics, but has not been shown to improve outcomes as adjunctive therapy. Combined antagonism has been tried only in single-dose studies in stable patients or over the short-term in acute heart failure, with encouraging results. Based on the both the pathophysiologic rationale for additional neurohormonal blockade and these results, chronically blocking both receptors, particularly in more congested patients, may offer significant benefit either as adjunctive or alternative therapy to standard diuretics.


Antidiuretic Hormone Receptor Antagonists/therapeutic use , Heart Failure , Hormone Replacement Therapy/methods , Receptors, Vasopressin/drug effects , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/physiopathology , Humans
17.
Eur J Heart Fail ; 20(4): 677-686, 2018 04.
Article En | MEDLINE | ID: mdl-29143416

AIMS: The direct renin inhibitor, aliskiren, is known to reduce plasma renin activity (PRA), but whether the efficacy of aliskiren varies based on an individual's baseline PRA in patients hospitalized for heart failure (HF) is presently unknown. We characterized the prognostic value of PRA and determined if this risk is modifiable with use of aliskiren. METHODS AND RESULTS: This pre-specified neurohormonal substudy of ASTRONAUT analysed all patients hospitalized for HF with ejection fraction (EF) ≤40% with available baseline PRA data (n = 1306, 80.9%). Risk associated with baseline PRA and short-term changes in PRA from baseline to 1 month was modelled with respect to 12-month clinical events. Median baseline PRA was 3.0 (interquartile range 0.6-16.4) ng/mL/h. Aliskiren significantly reduced PRA early after treatment initiation through 12-month follow-up compared with placebo (P < 0.001). The lowest baseline PRA quartile (<0.6 ng/mL/h) was independently predictive of lower all-cause mortality [adjusted hazard ratio (HR) 0.50, 95% confidence interval (CI) 0.31-0.81] and the composite of cardiovascular mortality and HF hospitalization (adjusted HR 0.57, 95% CI 0.40-0.79). Delta log-normalized PRA (from baseline to 1 month) was not predictive of either primary endpoint at 12 months (P ≥ 0.43). The prognostic value of baseline PRA and short-term changes in PRA did not vary by randomization to aliskiren or placebo (interaction P ≥ 0.13). CONCLUSIONS: Plasma renin activity is reduced early and durably by aliskiren, but this did not translate into improved clinical outcomes in ASTRONAUT. Baseline PRA or short-term reduction in PRA do not identify a subgroup who may preferentially benefit from direct renin inhibition. Clinical Trial Registration ClinicalTrials.gov Unique Identifier: NCT00894387.


Amides/administration & dosage , Fumarates/administration & dosage , Heart Failure/drug therapy , Inpatients , Renin-Angiotensin System/drug effects , Renin/blood , Aged , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Heart Failure/blood , Heart Failure/physiopathology , Humans , Male , Middle Aged , Prospective Studies , Renin/drug effects , Stroke Volume/physiology , Treatment Outcome
18.
Cardiol Clin ; 36(1): 63-72, 2018 Feb.
Article En | MEDLINE | ID: mdl-29173682

The majority of patients hospitalized with acute heart failure (AHF) initially present to the emergency department (ED). Correct diagnosis followed by prompt treatment ensures optimal outcomes. Paradoxically, identification of high risk is not the unmet need, given nearly all ED AHF patients are hospitalized; rather, it is identification of low-risk. Currently, no risk-stratification instrument can be universally recommended to safely discharge ED patients. With the exception of diagnosis, management recommendations are largely expert opinion, informed by existing evidence and tradition. In the absence of robust evidence, we propose a framework for management to guide the busy clinician.


Disease Management , Dyspnea , Emergency Service, Hospital , Heart Failure , Hemodynamics/physiology , Acute Disease , Dyspnea/etiology , Dyspnea/physiopathology , Dyspnea/therapy , Heart Failure/complications , Heart Failure/physiopathology , Heart Failure/therapy , Humans
19.
Eur J Heart Fail ; 20(4): 630-641, 2018 04.
Article En | MEDLINE | ID: mdl-29082676

Acute heart failure (HF) is a global pandemic with more than one million admissions to hospital annually in the US and millions more worldwide. Post-discharge mortality and readmission rates remain unchanged and unacceptably high. Although recent drug development programmes have failed to deliver novel therapies capable of reducing cardiovascular morbidity and mortality in patients hospitalized for worsening chronic HF, hospitalized HF registries and clinical trial databases have generated a wealth of information improving our collective understanding of the HF syndrome. This review will summarize key insights from clinical trials in acute HF and hospitalized HF registries over the last several decades, focusing on improving the management of patients with HF and reduced ejection fraction.


Heart Failure/epidemiology , Hospitalization/trends , Registries , Acute Disease , Global Health , Hospital Mortality/trends , Humans , Morbidity/trends , Prognosis , Survival Rate/trends
20.
Eur J Heart Fail ; 20(2): 281-291, 2018 02.
Article En | MEDLINE | ID: mdl-29044915

AIMS: Troponin levels are commonly elevated among patients hospitalized for heart failure (HF), but the prevalence and prognostic significance of early post-discharge troponin elevation are unclear. This study sought to describe the frequency and prognostic value of pre-discharge and post-discharge troponin elevation, including persistent troponin elevation from the inpatient to outpatient settings. METHODS AND RESULTS: The ASTRONAUT trial (NCT00894387; http://www.clinicaltrials.gov) enrolled hospitalized HF patients with ejection fraction ≤40% and measured troponin I prior to discharge (i.e. study baseline) and at 1-month follow-up in a core laboratory (elevation defined as >0.04 ng/mL). This analysis included 1469 (91.0%) patients with pre-discharge troponin data. Overall, 41.5% and 29.9% of patients had elevated pre-discharge [median: 0.09 ng/mL; interquartile range (IQR): 0.06-0.19 ng/mL] and 1-month (median: 0.09 ng/mL; IQR: 0.06-0.15 ng/mL) troponin levels, respectively. Among patients with pre-discharge troponin elevation, 60.4% had persistent elevation at 1 month. After adjustment, pre-discharge troponin elevation was not associated with 12-month clinical outcomes. In contrast, 1-month troponin elevation was independently predictive of increased all-cause mortality [hazard ratio (HR) 1.59, 95% confidence interval (CI) 1.18-2.13] and cardiovascular mortality or HF hospitalization (HR 1.28, 95% CI 1.03-1.58) at 12 months. Associations between 1-month troponin elevation and outcomes were similar among patients with newly elevated (i.e. normal pre-discharge) and persistently elevated levels (interaction P ≥ 0.16). The prognostic value of 1-month troponin elevation for 12-month mortality was driven by a pronounced association among patients with coronary artery disease (interaction P = 0.009). CONCLUSIONS: In this hospitalized HF population, troponin I elevation was common during index hospitalization and at 1-month follow-up. Elevated troponin I level at 1 month, but not pre-discharge, was independently predictive of increased clinical events at 12 months. Early post-discharge troponin I measurement may offer a practical means of risk stratification and should be investigated as a therapeutic target.


Heart Failure/blood , Patient Discharge/trends , Stroke Volume/physiology , Troponin/blood , Aged , Biomarkers/blood , Cause of Death/trends , Europe/epidemiology , Female , Follow-Up Studies , Heart Failure/mortality , Heart Failure/physiopathology , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Risk Factors , Survival Rate/trends , Time Factors , United States/epidemiology
...