1
|
Anker SD, Friede T, Butler J, Talha KM, Diek M, Nosko A, Placzek M, Hasenfuß G, Ponikowski P, Karakas M. Rationale and design of the FAIR-HF2-DZHK05 trial: Ferric carboxymaltose assessment of morbidity and mortality in patients with iron deficiency and chronic heart failure. Eur J Heart Fail 2025. [PMID: 39876699 DOI: 10.1002/ejhf.3574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
AIMS While it is widely accepted that intravenous (IV) iron improves functional capacity, symptoms, and cardiovascular outcomes in patients with heart failure (HF) with reduced ejection fraction (HFrEF) diagnosed with iron deficiency (ID), three recently published cardiovascular outcome trials (AFFIRM-AHF, IRONMAN and HEART-FID) of IV iron supplementation in HF failed to demonstrate a significant benefit on their respective primary endpoints. Dosing of IV iron after the initial correction of baseline ID - by design or as a result of trial circumstances - was relatively low (i.e. <500 mg/year). The primary objective of the FAIR-HF2 trial is to evaluate the treatment effect of ferric carboxymaltose (FCM) compared with placebo in ambulatory patients with HFrEF using a higher dose of IV iron during follow-up (i.e. >1000 mg/year). The second objective of the study is to create prospective evidence for patients fulfilling the new definition of ID for patients with HF, i.e. for those with a transferrin saturation <20%. METHODS FAIR-HF2 is an investigator-initiated, multicentre, randomized, double-blind, placebo-controlled trial that has recruited 1105 patients with chronic HF with a left ventricular ejection fraction of ≤45% and concomitant ID, defined as serum ferritin <100 ng/ml or serum ferritin 100-299 ng/ml with a transferrin saturation <20%. Patients were consented and randomized to receive either IV FCM (treatment) or saline (placebo). During an estimated median follow-up of over 2 years, patients underwent a placebo-controlled repletion and maintenance phase, with an initial iron supplementation of up to 2000 mg, followed by 500 mg every 4 months unless stop criteria of haemoglobin >16 mg/dl or serum ferritin >800 ng/ml are met on repeat visits. The trial will evaluate three primary hypotheses: (i) time to first event of cardiovascular death or hospitalization for HF, (ii) the rate of total (first and recurrent) HF hospitalizations (both analysed in the full study population), and (iii) the time to first event of cardiovascular death or hospitalization for HF in patients with a transferrin saturation <20% at baseline. The familywise type I error rate across the three primary endpoint hypotheses will be controlled using the Hochberg procedure (alpha 0.05). CONCLUSION The FAIR-HF2 will evaluate the efficacy of FCM in patients with HFrEF in improving cardiovascular outcomes by utilizing a more aggressive approach towards iron supplementation ensuring prevention of transitional ID after initial repletion targets have been met.
Collapse
Affiliation(s)
- Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité, Institute of Health Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, German Centre for Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Baylor Scott and White Research Institute, Dallas, TX, USA
| | - Khawaja M Talha
- Department of Cardiovascular Disease, Loyola University Medical Center, Maywood, IL, USA
| | - Monika Diek
- Department of Cardiology (CVK) of German Heart Center Charité, Institute of Health Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Anna Nosko
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Marius Placzek
- Department of Medical Statistics, University Medical Center Göttingen, German Centre for Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Gerd Hasenfuß
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, German Centre for Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Mahir Karakas
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
2
|
Kido K, Beavers CJ, Dulnuan K, Fida N, Guglin M, Ilonze OJ, Mentz RJ, Narang N, Rajagopalan N, Ramu B, Sattar Y, Sokos G, Jankowska EA. Management of Iron Deficiency in Heart Failure: Practical Considerations and Implementation of Evidence-Based Iron Supplementation. JACC. HEART FAILURE 2024; 12:1961-1978. [PMID: 39001744 DOI: 10.1016/j.jchf.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 07/15/2024]
Abstract
Iron deficiency (ID) is present in approximately 50% of patients with heart failure (HF) and even higher prevalence rate up to 80% in post-acute HF setting. The current guidelines for HF recommend intravenous (IV) iron replacement in HF with reduced or mildly reduced ejection fraction and ID based on clinical trials showing improvements in quality of life and exercise capacity, and an overall treatment benefit for recurrent HF hospitalization. However, several barriers cause challenges in implementing IV iron supplementation in practice due, in part, to clinician knowledge gaps and limited resource availability to protocolize routine utilization in appropriate patients. Thus, the current review will discuss practical considerations in ID treatment, implementation of evidence-based ID treatment to improve regional health disparities with toolkits, inclusion/exclusion criteria of IV iron supplementation, and clinical controversies in ID treatment, as well as gaps in evidence and questions to be answered.
Collapse
Affiliation(s)
- Kazuhiko Kido
- West Virginia University School of Pharmacy, Morgantown, West Virginia, USA.
| | - Craig J Beavers
- University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Kenneth Dulnuan
- Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | | | - Maya Guglin
- Krannert Cardiovascular Research Center, Indiana University Health School of Medicine, Indianapolis, Indiana, USA
| | - Onyedika J Ilonze
- Division of Cardiovascular Medicine, Krannert Cardiovascular Research Center, Indiana University, Indianapolis, Indiana, USA
| | - Robert J Mentz
- Duke University School of Medicine, Durham, North Carolina, USA
| | - Nikhil Narang
- Advocate Heart Institute, Oak Lawn, Illinois, USA; Division of Cardiology, Department of Medicine, University of Illinois-Chicago, Chicago, Illinois, USA
| | - Navin Rajagopalan
- Division of Cardiology, University of Kentucky, Lexington, Kentucky, USA
| | | | - Yasar Sattar
- West Virginia University Medicine, Morgantown, West Virginia, USA
| | - George Sokos
- West Virginia University Medicine, Morgantown, West Virginia, USA
| | | |
Collapse
|
3
|
Abdullah M, Abdo B, Ahmed F, Alzanen K, Albadani N, Badheeb M. Prevalence and independent predictors of Iron deficiency in Yemeni patients with congestive heart failure: a retrospective cross-sectional study. Sci Rep 2024; 14:28901. [PMID: 39572566 PMCID: PMC11582654 DOI: 10.1038/s41598-024-78556-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024] Open
Abstract
Heart failure (HF) constitutes a substantial burden on healthcare systems worldwide, contributing to elevated rates of mortality and morbidity. Iron deficiency (ID), both with and without concurrent anemia, has been identified in up to half of all patients with CHF and is associated with an increased risk of HF exacerbations, higher rates of hospitalization, and diminished quality of life. However, data from resource-limited settings remain limited. In this study, we reviewed 108 adult Yemeni patients with HF who also had concomitant ID, defined as serum ferritin concentrations of < 100 ng/mL or 100-299 ng/mL with transferrin saturation < 20%. The prevalence of ID among HF patients was determined, and independent predictors of ID were assessed using univariate and multivariate regression analyses. Anemia was present in 64 (59.3%), ID was observed in 65 (60.2%), and both anemia and ID were concurrently present in 44 (40.7%) patients. The mean ejection fraction among the study cohort was 34.2 ± 6.3%. Multivariate regression analysis identified New York Heart Association class III (OR: 4.46; 95% CI: 1.65-12.90, p = 0.004), presence of anemia (OR: 3.95; 95% CI: 1.51-11.23, p = 0.007), and an EF < 30% (OR: 9.42; 95% CI: 1.97-54.64, p = 0.007) as independent predictors of ID. These findings highlight the potential under-recognition of ID in patients with congestive HF, suggesting the need for routine assessment of iron status in this patient population.
Collapse
Affiliation(s)
- Mohammed Abdullah
- Department of Internal Medicine, School of Medicine, Ibb University, Ibb, Yemen
| | - Basheer Abdo
- Department of Internal Medicine, School of Medicine, Ibb University, Ibb, Yemen
| | - Faisal Ahmed
- Department of Urology, School of Medicine, Ibb University, Ibb, Yemen.
| | - Khaled Alzanen
- Department of Internal Medicine, School of Medicine, Ibb University, Ibb, Yemen
| | - Nabile Albadani
- Department of Internal Medicine, School of Medicine, Ibb University, Ibb, Yemen
| | - Mohamed Badheeb
- Internal Medicine, Yale New Haven Health, Bridgeport Hospital, Bridgeport, USA
| |
Collapse
|
4
|
Mohamed AA, Christensen DM, Mohammad M, Torp-Pedersen C, Køber L, Fosbøl EL, Biering-Sørensen T, Hansen ML, Malik ME, Nouhravesh N, Anderrson C, Schou M, Gislason G. Prognostic impact of iron deficiency in new-onset chronic heart failure: Danish Heart Failure Registry insights. ESC Heart Fail 2024. [PMID: 39505360 DOI: 10.1002/ehf2.15149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/12/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
AIMS Iron deficiency (ID) is prevalent in chronic heart failure (HF) but lacks a consensus definition. This study evaluates the prevalence and the prognostic impact of ID using different criteria on all-cause and cardiovascular mortality, as well as first hospitalization for HF in patients with new-onset chronic HF. METHODS In this nationwide registry-based cohort, we explored four definitions of ID: the current European Society of Cardiology (ESC) guidelines [ferritin <100 ng/mL or ferritin 100-299 ng/mL and transferrin saturation (TSAT) <20%], ferritin level <100 ng/mL, TSAT < 20% and serum iron ≤13 μmol/L. Patients were identified through the Danish Heart Failure Registry. RESULTS Of 9477 new-onset chronic HF patients registered in the Danish Heart Failure Registry from April 2003 to December 2019, we observed ID prevalence rates ranging from 35.8% to 64.3% depending on the ID definition used. Among patients with ID defined by iron ≤13 μmol/L or TSAT < 20%, 26% and 15.5%, respectively, did not meet the ESC guidelines definition for ID. Conversely, 11% of patients meeting the ESC criteria exhibited serum iron >13 μmol/L and TSAT > 20%. Regardless of anaemia status, ID defined by TSAT < 20% or serum iron ≤13 μmol/L was associated with all-cause mortality [non-anaemic, hazard ratio (HR): 1.57, 95% confidence interval (CI): 1.30-1.89 and HR: 1.47, 95% CI: 1.24-1.73; anaemic, HR: 1.22, 95% CI: 1.07-1.38 and HR: 1.25, 95% CI: 1.09-1.44, respectively] and cardiovascular mortality (non-anaemic, HR: 2.21, 95% CI: 1.59-3.06 and HR: 1.47, 95% CI: 1.12-1.95; anaemic, HR: 1.37, 95% CI: 1.11-1.69 and HR: 1.28, 95% CI: 1.02-1.61, respectively), as well as increased risk of first hospitalization for HF (non-anaemic, HR: 1.28, 95% CI: 1.09-1.1.50 and HR: 1.27, 95% CI: 1.10-1.46; anaemic, HR: 1.25, 95% CI: 1.08-1.44 and HR: 1.22, 95% CI: 1.05-1.42, respectively). ID defined by ESC guidelines was associated with all-cause and cardiovascular mortality only in non-anaemic patients (HR: 1.41, 95% CI: 1.18-1.1.70 and HR: 1.58, 95% CI: 1.18-2.12.). Furthermore, the ESC guideline definition was associated with increased risk of first hospitalization for HF, regardless of anaemia status (non-anaemic, HR: 1.26, 95% CI: 1.08-1.1.47; anaemic, HR: 1.34, 95% CI: 1.17-1.53). CONCLUSIONS ID, when defined by TSAT < 20% or serum iron ≤13 μmol/L, is associated with increased risk of all-cause and cardiovascular mortality, as well as first hospitalization for HF in patients with new-onset chronic HF, regardless of anaemia status. Conversely, ID defined as ESC guidelines is associated with all-cause and cardiovascular mortality only in non-anaemic patients.
Collapse
Affiliation(s)
- Abdullahi Ahmed Mohamed
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Daniel Mølager Christensen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Milan Mohammad
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Torp-Pedersen
- Department of Cardiology, Copenhagen University Hospital - Nordsjællands Hospital, Hillerød, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Emil Loldrup Fosbøl
- Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Tor Biering-Sørensen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Morten Lock Hansen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Mariam Elmegaard Malik
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Nina Nouhravesh
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Charlotte Anderrson
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Center for Advanced Heart Disease, Section of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Morten Schou
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Gunnar Gislason
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| |
Collapse
|
5
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
Ding Y, Zhang Y, Gao X, Hua C, Liu L, Huang D. Association of Iron Therapy with Mortality in Patients with Acute Myocardial Infarction and Iron Deficiency. Cardiovasc Toxicol 2024; 24:1018-1027. [PMID: 39093536 DOI: 10.1007/s12012-024-09905-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Iron deficiency (ID) is common in patients with acute myocardial infarction (AMI). It is unknown whether patients with AMI combined with ID will benefit from iron supplementation therapy. This study aimed to assess the relationship between iron therapy and mortality in AMI patients. Retrospective analysis was performed in subjects screened from the Medical Information Mart in Intensive Care-IV database. The data were obtained from ICU patients admitted to Beth Israel Deaconess Medical Center between 2008 and 2019. The patients were divided into two groups according to iron treatment exposure. Propensity score matching (PSM) was performed in the original cohort at a 1:1 ratio. Univariate and multivariate analyses were performed to adjust for confounding factors. The primary outcome was 28-day mortality. A total of 426 patients were included in this study. After 1:1 PSM, 208 patients were analyzed. Iron treatment was associated with a lower risk of 28-day mortality (9 deaths (8.65%) in the iron treatment group vs. 21 deaths (20.19%) in the non-iron treatment group; HR = 0.39; 95% CI = 0.17-0.89; p = 0.025) and in-hospital mortality (4 deaths (3.85%) in the iron treatment group vs. 12 deaths (11.54%) in the non-iron treatment group; OR, 0.15; 95% CI, 0.03-0.74; p = 0.029). Iron treatment was associated with reduced 28-day mortality in patients with AMI combined with ID. Iron treatment had no significant effect on the length of hospitalization or the length of ICU stay. Prospective studies are needed to verify this conclusion.
Collapse
Affiliation(s)
- Ye Ding
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yiyan Zhang
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xin Gao
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chang Hua
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Linsheng Liu
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Dan Huang
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
7
|
Beito M, Taegtmeyer H. Metabolic cycles: A unifying concept for energy transfer in the heart. J Mol Cell Cardiol 2024; 195:103-109. [PMID: 39154711 DOI: 10.1016/j.yjmcc.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
It is still debated whether changes in metabolic flux are cause or consequence of contractile dysfunction in non-ischemic heart disease. We have previously proposed a model of cardiac metabolism grounded in a series of six moiety-conserved, interconnected cycles. In view of a recent interest to augment oxygen availability in heart failure through iron supplementation, we integrated this intervention in terms of moiety conservation. Examining published work from both human and murine models, we argue this strategy restores a mitochondrial cycle of energy transfer by enhancing mitochondrial pyruvate carrier (MPC) expression and providing pyruvate as a substrate for carboxylation and anaplerosis. Metabolomic data from failing heart muscle reveal elevated pyruvate levels with a concomitant decrease in the levels of Krebs cycle intermediates. Additionally, MPC is downregulated in the same failing hearts, as well as under hypoxic conditions. MPC expression increases upon mechanical unloading in the failing human heart, as does contractile function. We note that MPC deficiency also alters expression of enzymes involved in pyruvate carboxylation and decarboxylation, increases intermediates of biosynthetic pathways, and eventually leads to cardiac hypertrophy and dilated cardiomyopathy. Collectively, we propose that an unbroken chain of moiety-conserved cycles facilitates energy transfer in the heart. We refer to the transport and subsequent carboxylation of pyruvate in the mitochondrial matrix as an example and a proposed target for metabolic support to reverse impaired contractile function.
Collapse
Affiliation(s)
- Mitchell Beito
- McGovern Medical School - The University of Texas Health Science Center at Houston, United States of America
| | - Heinrich Taegtmeyer
- McGovern Medical School - The University of Texas Health Science Center at Houston Department of Internal Medicine, Division of Cardiology, 6431 Fannin St. Houston, TX 77030, United States of America.
| |
Collapse
|
8
|
Ondrusek M, Artemiou P, Bezak B, Gasparovic I, By TMD, Durdik S, Lesny P, Goncalvesova E, Hulman M. Temporal Analysis in Outcomes of Long-Term Mechanical Circulatory Support: Retrospective Study. Thorac Cardiovasc Surg 2024; 72:521-529. [PMID: 38641334 DOI: 10.1055/s-0044-1782600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
BACKGROUND Mechanical assist device indications have changed in recent years. Reduced incidence of complications, better survival, and the third generation of mechanical support devices contributed to this change. In this single-center study, we focused on two time periods that are characterized by the use of different types of mechanical support devices, different patient characteristics, and change in the indications. METHODS The data were processed from the European Registry for Patients with Mechanical Circulatory Support (EUROMACS). We retrospectively defined two time intervals to reflect changes in ventricular assist device technology (period 1: 2007-2015; period 2: 2016-20222). A total of 181 patients underwent left ventricular assist device implantation. Device utilization was the following: HeartMate II = 52 (76.4%) and HeartWare = 16 (23.6%) in period 1 and HeartMate II = 2 (1.8%), HeartMate 3 = 70 (61:9%), HeartWare = 29 (25.7%), SynCardia TAH = 10 (8.8%), and BerlinHeart EXCOR = 2 (1.8%) in period 2. The outcomes of the time intervals were analyzed and evaluated. RESULTS Survival was significantly higher during the second time period. Multivariate analysis revealed that age and bypass pump time are independent predictors of mortality. Idiopathic cardiomyopathy, bypass time, and the Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS) score are independent predictors of adverse events. Furthermore, the first period was noted to be at an increased risk of the following adverse events: pump thrombosis, gastrointestinal bleeding, and bleeding events. CONCLUSION Despite the higher risk profile of the patients and persistent challenges, during the second period, there was a significant decrease in mortality and morbidity. The use of the HeartMate 3 device may have contributed to this result.
Collapse
Affiliation(s)
- Matej Ondrusek
- Faculty of Medicine of the Comenius University, National Institute of Cardiovascular diseases, Clinic of Cardiac Surgery, Bratislava, Slovakia
| | - Panagiotis Artemiou
- Faculty of Medicine of the Comenius University, National Institute of Cardiovascular diseases, Clinic of Cardiac Surgery, Bratislava, Slovakia
| | - Branislav Bezak
- Faculty of Medicine of the Comenius University, National Institute of Cardiovascular diseases, Clinic of Cardiac Surgery, Bratislava, Slovakia
| | - Ivo Gasparovic
- Faculty of Medicine of the Comenius University, National Institute of Cardiovascular diseases, Clinic of Cardiac Surgery, Bratislava, Slovakia
| | - Theo Mmh de By
- EUROMACS, European Association for Cardio-Thoracic Surgery (EACTS), Windsor, United Kingdom
| | - Stefan Durdik
- Faculty of Medicine, Comenius University, St. Elizabeth Oncology Institute, Clinic of Surgical Oncology, Bratislava, Slovakia
| | - Peter Lesny
- Faculty of Medicine of the Comenius University, National Institute of Cardiovascular Diseases, Clinic of Heart Failure, Bratislava, Slovakia
| | - Eva Goncalvesova
- Faculty of Medicine of the Comenius University, National Institute of Cardiovascular Diseases, Clinic of Heart Failure, Bratislava, Slovakia
| | - Michal Hulman
- Faculty of Medicine of the Comenius University, National Institute of Cardiovascular diseases, Clinic of Cardiac Surgery, Bratislava, Slovakia
| |
Collapse
|
9
|
Reza N, Pellicori P, Starling RC. REVOLUTION in Heart Failure Care and SELECT Highlights From the European Society of Cardiology-Heart Failure Association Heart Failure & World Congress on Acute Heart Failure 2024. J Card Fail 2024; 30:1157-1160. [PMID: 38838982 DOI: 10.1016/j.cardfail.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Affiliation(s)
- Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Pierpaolo Pellicori
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Randall C Starling
- Department of Cardiovascular Medicine, Kaufman Center for Heart Failure Treatment and Recovery, Heart, Vascular and Thoracic Institute, Cleveland, OH, USA
| |
Collapse
|
10
|
Wang M, Zhang D, Jiang L, Ye M, Nie J, Yin J. Association between serum transferrin saturation levels and heart failure in adults aged ≥40 years: a cross-sectional study based on NHANES (2017-2020.03). Front Endocrinol (Lausanne) 2024; 15:1419064. [PMID: 39280001 PMCID: PMC11392747 DOI: 10.3389/fendo.2024.1419064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Background Limited data are available regarding the association between serum transferrin saturation (TSAT) levels and heart failure (HF). Methods We utilized data from National Health and Nutrition Examination Survey (NHANES) 2017- 2020.03 for analysis. Data on TAST, HF and covariates were extracted and analyzed. Weighted logistic regression and subgroup analysis were used to explore the independent association between TSAT and HF. Furthermore, interaction tests were also carried out to evaluate the strata differences. We subsequently assessed whether there was a non-linear relationship between the 2 using Restricted cubic spline (RCS) and threshold effect models. Result A total of 282 (3.87%) participants were identified to have HF. Among the total population, participants with HF had significantly lower TSAT levels compared to those without HF (24.63 vs. 27.95, P = 0.001). After fully adjusting for potential confounders, weighted multiple logistic regression models revealed a 2.6% reduced in the risk of HF when each unit of TSAT increased. There was also a negative association between elevated TSAT and developed risk of HF in the quartile groups (Q1 OR:1.00; Q2 OR: 0.924 [95%CI:0.593,1.440]; Q3 OR: 0.515 [95%CI:0.298,0.891]; Q4 OR:0.411 [95%CI:0.201,0.839]). The subgroup analysis results remained consistent across strata, with a strong negative correlation between TSAT and HF. Interaction tests showed no dependence on gender, age, Body Mass Index, race, diabetes, hypertension, hyperlipidemia, ratio of family income to poverty and education for this negative association between TSAT and HF (all p for interaction >0.05). The RCS and threshold effect models indicated a linear negative correlation between TSAT and HF, which was more pronounced when TSAT under 40%. Conclusion Overall, these findings suggest a consistent and negative association between TSAT levels and the presence of HF among middle-aged and older adults in the United States.
Collapse
Affiliation(s)
- Mian Wang
- Department of Geriatric, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Dongyang Zhang
- Department of Geriatric, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Lanying Jiang
- Department of Geriatric, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Maosheng Ye
- Department of Geriatric, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Jing Nie
- Department of Geriatric, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Junjie Yin
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Zhang Y, Li B, Cai H, Fu Y, Zheng Y. Associations of iron metabolism and inflammation with all-cause and cardiovascular mortality in a large NHANES community sample: Moderating and mediating effects. Nutr Metab Cardiovasc Dis 2024; 34:1854-1863. [PMID: 38658228 DOI: 10.1016/j.numecd.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/01/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND AND AIMS This study aimed to assess the associations between serum iron concentration, C-reactive protein (CRP) concentration and the risk of all-cause mortality and cardiovascular mortality in the general population and to explore potential mediating and moderating effects. METHODS AND RESULTS This study analyzed data from the National Health and Nutrition Examination Survey spanning the years 1999-2010, encompassing 23,634 participants. Cox proportional hazards regression models were employed to investigate the independent associations of serum iron and CRP with all-cause and cardiovascular mortality. Moderation and mediation analyses explored the moderating effect of CRP on the association between the serum iron concentration and all-cause and cardiovascular mortality, and the mediating role of the serum iron concentration in the association between the CRP concentration and all-cause and cardiovascular mortality. After multivariate adjustments in the Cox model, serum iron and CRP levels were independently correlated with both all-cause and cardiovascular mortality risk. Moderation analyses revealed a more pronounced correlation between the serum iron concentration and both all-cause and cardiovascular mortality in participants with higher CRP levels. Mediation analysis indicated that the serum iron concentration partly mediated the impact of CRP on the risk of all-cause mortality (13.79%) and cardiovascular mortality (24.12%). CONCLUSION Serum iron and CRP are independently associated with all-cause and cardiovascular mortality. Moreover, the associations between serum iron concentrations and both all-cause and cardiovascular mortality are more pronounced in individuals with elevated CRP. Serum iron partially mediates the effect of CRP on all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Yaoting Zhang
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Bing Li
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - He Cai
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yu Fu
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yang Zheng
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
12
|
Jenča D, Melenovský V, Mrázková J, Šramko M, Kotrč M, Želízko M, Adámková V, Piťha J, Kautzner J, Wohlfahrt P. Iron deficiency and all-cause mortality after myocardial infarction. Eur J Intern Med 2024; 126:102-108. [PMID: 38697863 DOI: 10.1016/j.ejim.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Data on the clinical significance of iron deficiency (ID) in patients with myocardial infarction (MI) are conflicting. This may be related to the use of various ID criteria. We aimed to compare the association of different ID criteria with all-cause mortality after MI. METHODS Consecutive patients hospitalized for their first MI at a large tertiary heart center were included. We evaluated the association of different iron metabolism parameters measured on the first day after hospital admission with all-cause mortality. RESULTS From the 1,156 patients included (aged 64±12 years, 25 % women), 194 (16.8 %) patients died during the median follow-up of 3.4 years. After multivariate adjustment, iron level ≤13 µmol/L (HR 1.67, 95 % CI 1.19-2.34) and the combination of iron level ≤12.8 µmol/L and soluble transferrin receptor (sTfR) ≥3 mg/L (HR 2.56, 95 % CI 1.64-3.99) termed as PragueID criteria were associated with increased mortality risk and had additional predictive value to the GRACE score. Compared to the model including iron level, the addition of sTfR improved risk stratification (net reclassification improvement 0.61, 95 % CI 0.52-0.69) by reclassifying patients into a higher-risk group. No association between ferritin level and mortality was found. 51 % of patients had low iron levels, and 58 % fulfilled the PragueID criteria. CONCLUSION Iron deficiency is common among patients with the first MI. The PragueID criteria based on iron and soluble transferrin receptor levels provide the best prediction of mortality and should be evaluated in future interventional studies for the identification of patients potentially benefiting from intravenous iron therapy.
Collapse
Affiliation(s)
- Dominik Jenča
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic; Third Medical School, Charles University, Prague, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Jolana Mrázková
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Marek Šramko
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic; First Medical School, Charles University, Prague, Czech Republic
| | - Martin Kotrč
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Michael Želízko
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Věra Adámková
- Department of Preventive Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Piťha
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic; Medical and Dentistry School, Palacký University, Olomouc, Czech Republic
| | - Peter Wohlfahrt
- First Medical School, Charles University, Prague, Czech Republic; Department of Preventive Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| |
Collapse
|
13
|
Packer M, Anker SD, Butler J, Cleland JGF, Kalra PR, Mentz RJ, Ponikowski P, Talha KM. Critical re-evaluation of the identification of iron deficiency states and effective iron repletion strategies in patients with chronic heart failure. Eur J Heart Fail 2024; 26:1298-1312. [PMID: 38727791 DOI: 10.1002/ejhf.3237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/17/2024] [Accepted: 03/30/2024] [Indexed: 06/28/2024] Open
Abstract
According to current guidelines, iron deficiency is defined by a serum ferritin level <100 ng/ml or a transferrin saturation (TSAT) <20% if the serum ferritin level is 100-299 μg/L. These criteria were developed to encourage the use of intravenous iron as an adjunct to erythropoiesis-stimulating agents in the treatment of renal anaemia. However, in patients with heart failure, these criteria are not supported by any pathophysiological or clinical evidence that they identify an absolute or functional iron deficiency state. A low baseline TSAT-but not serum ferritin level-appears to be a reliable indicator of the effect of intravenous iron to reduce major heart failure events. In randomized controlled trials, intravenous iron decreased the risk of cardiovascular death or total heart failure hospitalization in patients with a TSAT <20% (risk ratio 0.67 [0.49-0.92]) but not in patients with a TSAT ≥20% (risk ratio 0.99 [0.74-1.30]), with the magnitude of the risk reduction being proportional to the severity of hypoferraemia. Patients who were enrolled in clinical trials solely because they had a serum ferritin level <100 μg/L showed no significant benefit on heart failure outcomes, and it is noteworthy that serum ferritin levels of 20-300 μg/L lie entirely within the range of normal values for healthy adults. Current guidelines reflect the eligibility criteria of clinical trials, which inadvertently adopted unvalidated criteria to define iron deficiency. Reliance on these guidelines would lead to the treatment of many patients who are not iron deficient (serum ferritin level <100 μg/L but normal TSAT) and ignores the possibility of iron deficiency in patients with a low TSAT but with serum ferritin level of >300 μg/L. Importantly, analyses of benefit based on trial eligibility-driven guidelines substantially underestimate the magnitude of heart-failure-event risk reduction with intravenous iron in patients who are truly iron deficient. Based on all available data, we recommend a new mechanism-based and trial-tested approach that reflects the totality of evidence more faithfully than the historical process adopted by clinical investigators and by the guidelines. Until additional evidence is forthcoming, an iron deficiency state in patients with heart failure should be defined by a TSAT <20% (as long as the serum ferritin level is <400 μg/L), and furthermore, the use of a serum ferritin level <100 μg/L alone as a diagnostic criterion should be discarded.
Collapse
Affiliation(s)
- Milton Packer
- Baylor University Medical Center, Dallas, TX, USA
- Imperial College, London, UK
| | - Stefan D Anker
- Department of Cardiology of German Heart Center Charité, Institute of Health Center for Regenerative Therapies, German Centre for Cardiovascular Research, Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Javed Butler
- Baylor Scott and White Research Institute, Baylor University Medical Center, Dallas, TX, USA
- University of Mississippi Medical Center, Jackson, MS, USA
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Paul R Kalra
- Department of Cardiology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
- Faculty of Science and Health, University of Portsmouth, Portsmouth, UK
| | - Robert J Mentz
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, and Duke Clinical Research Institute, Durham, NC, USA
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, Wroclaw, Poland
| | | |
Collapse
|
14
|
Fabbri M, Sahu A. Challenges and opportunities in patients with adult congenital heart disease, a narrative review. Front Cardiovasc Med 2024; 11:1366572. [PMID: 38873271 PMCID: PMC11171728 DOI: 10.3389/fcvm.2024.1366572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 05/10/2024] [Indexed: 06/15/2024] Open
Abstract
Adult congenital heart disease Pregnancy Transition of care Challenges heart failure.
Collapse
Affiliation(s)
- Matteo Fabbri
- Department of Cardiovascular Disease, Inova Heart and Vascular Institute, Falls Church, VA, United States
| | - Anurag Sahu
- Department of Cardiovascular Disease, Inova Heart and Vascular Institute, Falls Church, VA, United States
- Department of Cardiovascular Imaging, NIH/NHLBI Cardiovascular Imaging Lab, Bethesda, MD, United States
- Department of Cardiovascular Disease, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
15
|
Silva PX, Aguiar L, Gaspar M, Faustino P, Falcão LM, Barbosa M, Bicho M, Inácio Â. Analysis of Genes Involved in Oxidative Stress and Iron Metabolism in Heart Failure: A Step Forward in Risk Stratification. Cureus 2024; 16:e60707. [PMID: 38899268 PMCID: PMC11186693 DOI: 10.7759/cureus.60707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 06/21/2024] Open
Abstract
INTRODUCTION Heart failure (HF) is a clinical syndrome characterized by cardinal symptoms that may be accompanied by signs. It results from structural and/or functional abnormalities of the heart leading to elevated intracardiac pressures and/or inadequate cardiac output at rest and/or during exercise. The prevalence of iron deficiency and anemia justifies the current guidelines recommendation of screening. Genes HP, ACE, MTHFR, HFE, and CYBA are involved in oxidative mechanisms, iron metabolism, and hematologic homeostasis. This study investigates the contribution of variants Hp1/2 (HP), I/D (ACE), C677T (MTHFR), C282Y and H63D (HFE), and C242T (CYBA) to the development of HF, either independently or in epistasis. METHODS We used a database of 389 individuals, 143 HF patients, and 246 healthy controls. Genotypes were characterized through PAGE electrophoresis, PCR, PCR-RFLP, and multiplex-ARMS. Data analysis was performed with the SPSS® 26.0 software (IBM Corp., Armonk, NY). RESULTS We observed a significant association between the MTHFR gene and HF predisposition. The presence of allele T and genotype CT constituted risk, while genotype CC granted protection. Epistatic interactions revealed risk between genotype II of the ACE gene and genotypes CC (C282Y) or HH (H63D) of the HFE gene. Risk was also observed for interactions between genotype CC (CYBA)and genotypes 2-2 (HP), CT (MTHFR), or HH (HFE-H63D). CONCLUSION We concluded that genes HP, ACE, MTHFR, HFE, and CYBA contribute to the susceptibility for HF, individually or in epistasis. This study contributes to the clarification of the role that genes involved in oxidative mechanisms and iron metabolism play in the physiopathology of HF. It is, therefore, a step forward in risk stratification and personalized medicine.
Collapse
Affiliation(s)
- Pedro X Silva
- Genetics Laboratory, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Associate Laboratory TERRA, Environmental Health Institute, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
| | - Laura Aguiar
- Associate Laboratory TERRA, Environmental Health Institute, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Department of Genetics, Bento da Rocha Cabral Institute for Scientific Research, Lisbon, PRT
| | - Marcos Gaspar
- Genetics Laboratory, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Associate Laboratory TERRA, Environmental Health Institute, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Department of Genetics, Bento da Rocha Cabral Institute for Scientific Research, Lisbon, PRT
| | - Paula Faustino
- Department of Human Genetics, National Institute of Health Dr. Ricardo Jorge, Lisbon, PRT
- Associate Laboratory TERRA, Environmental Health Institute, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
| | - Luiz M Falcão
- Department of Genetics, Bento da Rocha Cabral Institute for Scientific Research, Lisbon, PRT
- Cardiovascular Centre of the University of Lisbon (CCUL@RISE), Faculty of Medicine of the University of Lisbon, Lisbon, PRT
| | - Mário Barbosa
- Department of Medicine, Hospital Lusíadas Lisboa, Lisbon, PRT
| | - Manuel Bicho
- Genetics Laboratory, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Associate Laboratory TERRA, Environmental Health Institute, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Department of Genetics, Bento da Rocha Cabral Institute for Scientific Research, Lisbon, PRT
| | - Ângela Inácio
- Genetics Laboratory, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Associate Laboratory TERRA, Environmental Health Institute, Faculty of Medicine of the University of Lisbon, Lisbon, PRT
- Department of Genetics, Bento da Rocha Cabral Institute for Scientific Research, Lisbon, PRT
| |
Collapse
|
16
|
Tsangaris A, Ambrosy AP, Tschida M, Alexy T. Settling the IRONy of Anemia in Heart Failure: Current Evidence and Future Directions. J Card Fail 2024; 30:691-693. [PMID: 38401670 DOI: 10.1016/j.cardfail.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Affiliation(s)
| | - Andrew P Ambrosy
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA; Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | | | - Tamas Alexy
- Division of Cardiology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
17
|
Gale SE, Willeford A, Sandquist K, Watson K. Intravenous iron in patients with iron deficiency and heart failure: a review of modern evidence. Curr Opin Cardiol 2024; 39:178-187. [PMID: 38353280 DOI: 10.1097/hco.0000000000001121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW Iron deficiency is common in patients with heart failure, affecting up to half of ambulatory patients and an even greater percentage of patients admitted for acute decompensation. Iron deficiency in this population is also associated with poor outcomes, including worse quality of life in addition to increased hospitalizations for heart failure and mortality. Evidence suggests that patients with iron deficiency in heart failure may benefit from repletion with IV iron. RECENT FINDINGS In this review, we outline the etiology and pathophysiology of iron deficiency in heart failure as well as various iron formulations available. We discuss evidence for intravenous iron repletion with a particular focus on recent studies that have evaluated its effects on hospitalizations and mortality. Finally, we discuss areas of uncertainty and future study and provide practical guidance for iron repletion. SUMMARY In summary, there is overwhelming evidence that intravenous iron repletion in patients with iron deficiency in heart failure is both beneficial and safe. However, further evidence is needed to better identify which patients would most benefit from iron repletion as well as the ideal repletion strategy.
Collapse
Affiliation(s)
- Stormi E Gale
- Novant Health Heart and Vascular Institute, Huntersville, North Carolina
| | - Andrew Willeford
- University of California San Diego Skaggs School of Pharmacy and Pharmaceutical Sciences, San Diego, California
| | | | - Kristin Watson
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Perillo A, Basile C, Fucile I, Rozza F, De Luca N, Mancusi C. Lung ultrasound at discharge predicts outcomes in heart failure: a pilot study. J Cardiovasc Med (Hagerstown) 2024; 25:394-396. [PMID: 38526933 PMCID: PMC10990019 DOI: 10.2459/jcm.0000000000001613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/07/2024] [Accepted: 03/01/2024] [Indexed: 03/27/2024]
Affiliation(s)
- Andrea Perillo
- Department of Advanced Biomedical Science, Federico II University Hospital, Naples, Italy
| | | | | | | | | | | |
Collapse
|
19
|
Sheehan M, Sokoloff L, Reza N. Acute Heart Failure: From The Emergency Department to the Intensive Care Unit. Cardiol Clin 2024; 42:165-186. [PMID: 38631788 PMCID: PMC11064814 DOI: 10.1016/j.ccl.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Acute heart failure (AHF) is a frequent cause of hospitalization around the world and is associated with high in-hospital and post-discharge morbidity and mortality. This review summarizes data on diagnosis and management of AHF from the emergency department to the intensive care unit. While more evidence is needed to guide risk stratification and care of patients with AHF, hospitalization is a key opportunity to optimize evidence-based medical therapy for heart failure. Close linkage to outpatient care is essential to improve post-hospitalization outcomes.
Collapse
Affiliation(s)
- Megan Sheehan
- Division of Internal Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Maloney Building 5th Floor, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Lara Sokoloff
- Division of Internal Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Maloney Building 5th Floor, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, 11th Floor South Pavilion, Room 11-145, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Sun Y, Peng W, Lin S, Cui J, Lu J. Iron Metabolic Biomarkers and the Mortality Risk in the General Population: A Nationwide Population-Based Cohort Study. J Endocr Soc 2024; 8:bvae063. [PMID: 38623382 PMCID: PMC11017327 DOI: 10.1210/jendso/bvae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Indexed: 04/17/2024] Open
Abstract
Context Iron is an essential element in the human body and plays a critical role in many physiological and cellular processes. However, the association between iron status and the risk of all-cause or cause-specific mortality has not been well-investigated. And it is unclear whether the association between iron metabolic biomarkers and the risk of mortality differs between people with and without diabetes mellitus (DM). Objective This work aimed to investigate associations between iron metabolic biomarkers and all-cause and cause-specific mortality risk in the general population, and heterogeneities in the associations among population with and without DM.. Methods A total of 29 166 adults from the National Health and Nutrition Examination Survey (NHANES) III and NHANES 1999 to 2010 were included, with linkage to the National Death Index to December 31, 2019. Cox proportional-hazard models and Fine-Gray subdistribution hazard models were used to estimate associations between iron metabolic biomarkers and outcomes. Results During a median follow-up of 18.83 years, 9378 deaths were observed, including 3420 cardiovascular disease (CVD) deaths and 1969 cancer deaths. A significant linear association between serum ferritin (SF) and all-cause mortality was observed among the overall population and those without DM. J-shaped associations between transferrin saturation (TSAT) and all-cause and CVD mortality were observed among all populations. In the overall population, compared to the first quartile (Q1) group, the adjusted hazard ratio (HR) (95% CI) for all-cause mortality was 1.07 (1.00-1.15), 1.05 (0.98-1.12), 1.13 (1.05-1.21) in Q2, Q3, and Q4 groups for SF, while the HR was 0.94 (0.88-0.99), 0.92 (0.86-0.97), and 0.93 (0.88-0.99) for TSAT. In individuals without DM, the adjusted HR of the Q4 of SF were 1.19 (1.03-1.37) for CVD mortality and 1.25 (1.05-1.48) for cancer mortality. In individuals with DM, the adjusted HRs of the Q4 of TSAT were 0.76 (0.62-0.93) for CVD mortality and 1.47 (1.07-2.03) for cancer mortality. Conclusion Iron metabolism abnormalities increase mortality risk in the general population. The associations of iron status with mortality were significantly different between individuals with and without DM, which indicated tailored strategies for iron homeostasis are needed.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Wenyao Peng
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Siqi Lin
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Jingjing Cui
- Department of Geriatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Jiapeng Lu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| |
Collapse
|
21
|
Thankachen J, Hall S, Alam A. Iron therapy for patients with iron deficiency and heart failure: a treatment strategy that is increasingly opaque. Proc AMIA Symp 2024; 37:477-478. [PMID: 38628330 PMCID: PMC11018066 DOI: 10.1080/08998280.2024.2329010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024] Open
Affiliation(s)
- Jincy Thankachen
- Division of Cardiology, New York University Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Shelley Hall
- Center for Advanced Heart and Lung Disease, Baylor University Medical Center, Baylor Scott and White Health, Dallas, Texas, USA
| | - Amit Alam
- Division of Cardiology, New York University Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| |
Collapse
|
22
|
Butler J, Mentz RJ, Hernandez AF. Iron replacement therapy in heart failure: Contextualizing the results of the HEART-FID trial. Eur J Heart Fail 2024. [PMID: 38439623 DOI: 10.1002/ejhf.3193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Affiliation(s)
- Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- University of Mississippi, Jackson, MS, USA
| | | | | |
Collapse
|
23
|
Gale SE. Extending the Evidence for Intravenous Iron in Patients With Heart Failure and Iron Deficiency. JACC. HEART FAILURE 2024; 12:537-538. [PMID: 38206236 DOI: 10.1016/j.jchf.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024]
Affiliation(s)
- Stormi E Gale
- Heart and Vascular Institute, Novant Health, Huntersville, North Carolina, USA.
| |
Collapse
|
24
|
Alagiakrishnan K, Mah D, Aronow WS, Lam PH, Frishman WH, Ahmed A, Deedwania P. Considerations Regarding Management of Heart Failure in Older Adults. Cardiol Rev 2024:00045415-990000000-00223. [PMID: 38421170 DOI: 10.1097/crd.0000000000000677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Understanding noncardiovascular comorbidities and geriatric syndromes in elderly patients with heart failure (HF) is important as the average age of the population increases. Healthcare professionals need to consider these complex dynamics when managing older adults with HF, especially those older than 80. A number of small studies have described associations between HF and major geriatric domains. With information on patients' cognitive, functional decline, and ability to adhere to therapy, physicians can plan for individualized treatment goals and recommendations for these patients.
Collapse
Affiliation(s)
| | - Darren Mah
- Department of Radiology, University of Alberta, Edmonton, Alberta, Canada
| | - Wilbert S Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Phillip H Lam
- Department of Medicine, Georgetown University, MedStar Washington Hospital Center, Washington, DC
| | | | - Ali Ahmed
- Center for Data Science and Outcomes Research, Veterans Affairs Medical Center, George Washington University, Georgetown University, Washington, DC; and
| | | |
Collapse
|
25
|
Youmans QR, Lala A, Mentz RJ. JCF Heart Failure Year-In-Review 2023… Shaping the Future of Heart Failure. J Card Fail 2024; 30:1-3. [PMID: 38212089 DOI: 10.1016/j.cardfail.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Affiliation(s)
- Quentin R Youmans
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anuradha Lala
- Zena and Michael A. Wiener Cardiovascular Institute and Department of Population Health Science and Policy, Mount Sinai, New York, NY, USA
| | - Robert J Mentz
- Duke University Medical Center and Duke Clinical Research Institute, Durham, NC, USA
| |
Collapse
|
26
|
Uskach TM, Tereschenko SN. [The Effects of Therapy for Iron Deficiency in Patients With Different Etiologies of Heart Failure and Concomitant Diseases]. KARDIOLOGIIA 2023; 63:87-95. [PMID: 38088117 DOI: 10.18087/cardio.2023.11.n2604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 12/18/2023]
Abstract
Iron deficiency (ID) in patients with heart failure (HF) is a factor for unfavorable course and prognosis of the disease. The significance of ID in the diagnosis and treatment of HF has previously been demonstrated by multiple studies and meta-analyses. Therapy for ID in patients with HF is one of the most relevant and discussed issues. The use of intravenous iron medicinal products for the treatment of ID is currently being actively studied in patients of various categories; attempts are being made to specify the indications for use to produce the greatest effect on the prognosis and quality of life of HF patients.
Collapse
Affiliation(s)
- T M Uskach
- Chazov National Medical Research Center, Moscow; Russian Medical Academy of Continuous Professional Education, Moscow
| | | |
Collapse
|
27
|
Anker SD, Filippatos G, Anker MS. Ferric Carboxymaltose in Heart Failure with Iron Deficiency. N Engl J Med 2023; 389:10.1056/NEJMc2311874#sa3. [PMID: 38048198 DOI: 10.1056/nejmc2311874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
|
28
|
Mentz RJ, Garg J, Rockhold FW, Butler J, De Pasquale CG, Ezekowitz JA, Lewis GD, O'Meara E, Ponikowski P, Troughton RW, Wong YW, She L, Harrington J, Adamczyk R, Blackman N, Hernandez AF. Ferric Carboxymaltose in Heart Failure with Iron Deficiency. N Engl J Med 2023; 389:975-986. [PMID: 37632463 DOI: 10.1056/nejmoa2304968] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/28/2023]
Abstract
BACKGROUND Ferric carboxymaltose therapy reduces symptoms and improves quality of life in patients who have heart failure with a reduced ejection fraction and iron deficiency. Additional evidence about the effects of ferric carboxymaltose on clinical events is needed. METHODS In this double-blind, randomized trial, we assigned ambulatory patients with heart failure, a left ventricular ejection fraction of 40% or less, and iron deficiency, in a 1:1 ratio, to receive intravenous ferric carboxymaltose or placebo, in addition to standard therapy for heart failure. Ferric carboxymaltose or placebo was given every 6 months as needed on the basis of iron indexes and hemoglobin levels. The primary outcome was a hierarchical composite of death within 12 months after randomization, hospitalizations for heart failure within 12 months after randomization, or change from baseline to 6 months in the 6-minute walk distance. The significance level was set at 0.01. RESULTS We enrolled 3065 patients, of whom 1532 were randomly assigned to the ferric carboxymaltose group and 1533 to the placebo group. Death by month 12 occurred in 131 patients (8.6%) in the ferric carboxymaltose group and 158 (10.3%) in the placebo group; a total of 297 and 332 hospitalizations for heart failure, respectively, occurred by month 12; and the mean (±SD) change from baseline to 6 months in the 6-minute walk distance was 8±60 and 4±59 m, respectively (Wilcoxon-Mann-Whitney P = 0.02; unmatched win ratio, 1.10; 99% confidence interval, 0.99 to 1.23). Repeated dosing of ferric carboxymaltose appeared to be safe with an acceptable adverse-event profile in the majority of patients. The number of patients with serious adverse events occurring during the treatment period was similar in the two groups (413 patients [27.0%] in the ferric carboxymaltose group and 401 [26.2%] in the placebo group). CONCLUSIONS Among ambulatory patients who had heart failure with a reduced ejection fraction and iron deficiency, there was no apparent difference between ferric carboxymaltose and placebo with respect to the hierarchical composite of death, hospitalizations for heart failure, or 6-minute walk distance. (Funded by American Regent, a Daiichi Sankyo Group company; HEART-FID ClinicalTrials.gov number, NCT03037931.).
Collapse
Affiliation(s)
- Robert J Mentz
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Jyotsna Garg
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Frank W Rockhold
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Javed Butler
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Carmine G De Pasquale
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Justin A Ezekowitz
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Gregory D Lewis
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Eileen O'Meara
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Piotr Ponikowski
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Richard W Troughton
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Yee Weng Wong
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Lilin She
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Josephine Harrington
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Robert Adamczyk
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Nicole Blackman
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| | - Adrian F Hernandez
- From the Division of Cardiology, Department of Medicine (R.J.M., J.H., A.F.H.), and the Department of Biostatistics and Bioinformatics (F.W.R.), Duke University School of Medicine, and Duke Clinical Research Institute (R.J.M., J.G., F.W.R., L.S., J.H., A.F.H.) - both in Durham, NC; Baylor Scott and White Research Institute, Dallas (J.B.); the Department of Medicine, University of Mississippi, Jackson (J.B.); Flinders Medical Centre, Flinders University, Adelaide, SA (C.G.D.P.), and the Department of Cardiology, Prince Charles Hospital and Faculty of Medicine, University of Queensland, Brisbane (Y.W.W.) - both in Australia; Canadian VIGOUR Centre, University of Alberta, Edmonton (J.A.E.), and Montreal Heart Institute and Université de Montréal, Montreal (E.O.) - both in Canada; the Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston (G.D.L.); the Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland (P.P.); Christchurch Heart Institute, University of Otago, Christchurch, New Zealand (R.W.T.); the Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (Y.W.W.); and American Regent, Shirley, NY (R.A., N.B.)
| |
Collapse
|