101
|
Kalra PR, Cleland JGF, Petrie MC, Thomson EA, Kalra PA, Squire IB, Ahmed FZ, Al-Mohammad A, Cowburn PJ, Foley PWX, Graham FJ, Japp AG, Lane RE, Lang NN, Ludman AJ, Macdougall IC, Pellicori P, Ray R, Robertson M, Seed A, Ford I. Intravenous ferric derisomaltose in patients with heart failure and iron deficiency in the UK (IRONMAN): an investigator-initiated, prospective, randomised, open-label, blinded-endpoint trial. Lancet 2022; 400:2199-2209. [PMID: 36347265 DOI: 10.1016/s0140-6736(22)02083-9] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND For patients with heart failure, reduced left ventricular ejection fraction and iron deficiency, intravenous ferric carboxymaltose administration improves quality of life and exercise capacity in the short-term and reduces hospital admissions for heart failure up to 1 year. We aimed to evaluate the longer-term effects of intravenous ferric derisomaltose on cardiovascular events in patients with heart failure. METHODS IRONMAN was a prospective, randomised, open-label, blinded-endpoint trial done at 70 hospitals in the UK. Patients aged 18 years or older with heart failure (left ventricular ejection fraction ≤45%) and transferrin saturation less than 20% or serum ferritin less than 100 μg/L were eligible. Participants were randomly assigned (1:1) using a web-based system to intravenous ferric derisomaltose or usual care, stratified by recruitment context and trial site. The trial was open label, with masked adjudication of the outcomes. Intravenous ferric derisomaltose dose was determined by patient bodyweight and haemoglobin concentration. The primary outcome was recurrent hospital admissions for heart failure and cardiovascular death, assessed in all validly randomly assigned patients. Safety was assessed in all patients assigned to ferric derisomaltose who received at least one infusion and all patients assigned to usual care. A COVID-19 sensitivity analysis censoring follow-up on Sept 30, 2020, was prespecified. IRONMAN is registered with ClinicalTrials.gov, NCT02642562. FINDINGS Between Aug 25, 2016, and Oct 15, 2021, 1869 patients were screened for eligibility, of whom 1137 were randomly assigned to receive intravenous ferric derisomaltose (n=569) or usual care (n=568). Median follow-up was 2·7 years (IQR 1·8-3·6). 336 primary endpoints (22·4 per 100 patient-years) occurred in the ferric derisomaltose group and 411 (27·5 per 100 patient-years) occurred in the usual care group (rate ratio [RR] 0·82 [95% CI 0·66 to 1·02]; p=0·070). In the COVID-19 analysis, 210 primary endpoints (22·3 per 100 patient-years) occurred in the ferric derisomaltose group compared with 280 (29·3 per 100 patient-years) in the usual care group (RR 0·76 [95% CI 0·58 to 1·00]; p=0·047). No between-group differences in deaths or hospitalisations due to infections were observed. Fewer patients in the ferric derisomaltose group had cardiac serious adverse events (200 [36%]) than in the usual care group (243 [43%]; difference -7·00% [95% CI -12·69 to -1·32]; p=0·016). INTERPRETATION For a broad range of patients with heart failure, reduced left ventricular ejection fraction and iron deficiency, intravenous ferric derisomaltose administration was associated with a lower risk of hospital admissions for heart failure and cardiovascular death, further supporting the benefit of iron repletion in this population. FUNDING British Heart Foundation and Pharmacosmos.
Collapse
Affiliation(s)
- Paul R Kalra
- Department of Cardiology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK; College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Faculty of Science and Health, University of Portsmouth, Portsmouth, UK
| | - John G F Cleland
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | | | - Philip A Kalra
- Department of Renal Medicine, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Fozia Z Ahmed
- Department of Cardiology, Manchester University NHS Foundation Trust, Manchester, UK
| | - Abdallah Al-Mohammad
- Department of Cardiology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, UK
| | - Peter J Cowburn
- Department of Cardiology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Paul W X Foley
- Wiltshire Cardiac Centre, Great Western Hospitals NHS Foundation Trust, Swindon, UK
| | - Fraser J Graham
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Alan G Japp
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Rebecca E Lane
- Department of Cardiology, Royal Brompton and Harefield Hospitals, London, UK
| | - Ninian N Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew J Ludman
- Department of Cardiology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | | | - Pierpaolo Pellicori
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Robin Ray
- Cardiology Department, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Michele Robertson
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Alison Seed
- Department of Cardiology, Blackpool Teaching Hospitals NHS Foundation Trust, Blackpool, UK
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK.
| |
Collapse
|
102
|
Lupu M, Tudor D, Filip A. Iron metabolism and cardiovascular disease: Basic to translational purviews and therapeutical approach. Rev Port Cardiol 2022; 41:1037-1046. [PMID: 36228833 DOI: 10.1016/j.repc.2021.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/09/2021] [Accepted: 09/13/2021] [Indexed: 11/15/2022] Open
Abstract
Iron interactions with the cardiovascular system were proposed about half a century ago, yet a clear-cut understanding of this micronutrient and its intricacies with acute and chronic events is still lacking. In chronic heart failure, patients with decreased iron stores appear to benefit from intravenous administration of metallic formulations, whereas acute diseases (e.g., myocardial infarction, stroke) are barely studied in randomized controlled trials in humans. However, proof-of-concept studies have indicated that the dual redox characteristics of iron could be involved in atherosclerosis, necrosis, and ferroptosis. To this end, we sought to review the currently available body of literature pertaining to these temporal profiles of heart diseases, as well as the pathophysiologic mechanism by which iron enacts, underlining key points related to treatment options.
Collapse
Affiliation(s)
- Mihai Lupu
- Iuliu Hatieganu University of Medicine and Pharmacy, Department of Physiology, Cluj-Napoca, Romania.
| | - Diana Tudor
- Iuliu Hatieganu University of Medicine and Pharmacy, Department of Physiology, Cluj-Napoca, Romania
| | - Adriana Filip
- Iuliu Hatieganu University of Medicine and Pharmacy, Department of Physiology, Cluj-Napoca, Romania
| |
Collapse
|
103
|
Martens P, Tang WHW. Iron Deficiency in Heart Failure and Pulmonary Hypertension. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2022; 24:213-229. [PMID: 38994176 PMCID: PMC11238656 DOI: 10.1007/s11936-022-00971-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/15/2022] [Indexed: 11/24/2022]
Abstract
Purpose of review To describe the role of iron deficiency in both heart failure and pulmonary hypertension. Recent findings To role of iron deficiency in heart failure is well established and pathophysiologic overlap with pulmonary hypertension exists. Summary Iron deficiency is common co-morbidity in heart failure and pulmonary hypertension. The high prevalence is intertwined into the pathophysiology of these conditions (e.g., neurohormonal activation, inflammation). The presence of iron deficiency has a negative impact on cardiomyocytes and cardiac function, skeletal muscle function, and pulmonary vascular function. In heart failure data from over 2000 randomized patients with iron deficiency using a uniform diagnosis, have illustrated beneficial effects on functional status, quality of life, reverse cardiac remodeling, and heart failure admissions. While iron deficiency is recognized to be prevalent in pulmonary hypertension and associated with worse functional status, the absence of a uniform definition and the absence of large prospective randomized controlled trials with iron therapies limits the conclusions on the causal role of iron deficiency such as observed in heart failure.
Collapse
Affiliation(s)
- Pieter Martens
- Department of Cardiovascular Medicine, Kauffman Center for Heart Failure Treatment and Recovery, Heart, Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Kauffman Center for Heart Failure Treatment and Recovery, Heart, Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
104
|
Management of Iron Deficiency in Heart Failure: A Review of Evidence. J Cardiovasc Pharmacol 2022; 80:755-768. [PMID: 36027595 DOI: 10.1097/fjc.0000000000001341] [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] [Received: 06/17/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Iron deficiency is common in patients with heart failure and has been associated with worse outcomes, including increases in mortality, disease progression, and hospitalizations. As such, several studies have evaluated the role of iron supplementation in mitigating these risks. Evidence for the role of intravenous iron in improving exercise capacity, quality of life, and hospitalizations is promising, although the benefits of oral iron remain less clear. This review will evaluate the literature surrounding iron supplementation in heart failure and provide practical recommendations for its management.
Collapse
|
105
|
Packer M. How can sodium-glucose cotransporter 2 inhibitors stimulate erythrocytosis in patients who are iron-deficient? Implications for understanding iron homeostasis in heart failure. Eur J Heart Fail 2022; 24:2287-2296. [PMID: 36377108 PMCID: PMC10100235 DOI: 10.1002/ejhf.2731] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 11/17/2022] Open
Abstract
Many patients with heart failure have an iron-deficient state, which can limit erythropoiesis in erythroid precursors and ATP production in cardiomyocytes. Yet, treatment with sodium-glucose cotransporter 2 (SGLT2) inhibitors produces consistent increases in haemoglobin and haematocrit, even in patients who are iron-deficient before treatment, and this effect remains unattenuated throughout treatment even though SGLT2 inhibitors further aggravate biomarkers of iron deficiency. Heart failure is often accompanied by systemic inflammation, which activates hepcidin, thus impairing the duodenal absorption of iron and the release of iron from macrophages and hepatocytes, leading to a decline in circulating iron. Inflammation and oxidative stress also promote the synthesis of ferritin and suppress ferritinophagy, thus impairing the release of intracellular iron stores and leading to the depletion of bioreactive cytosolic Fe2+ . By alleviating inflammation and oxidative stress, SGLT2 inhibitors down-regulate hepcidin, upregulate transferrin receptor protein 1 and reduce ferritin; the net result is to increase the levels of cytosolic Fe2+ available to mitochondria, thus enabling the synthesis of heme (in erythroid precursors) and ATP (in cardiomyocytes). The finding that SGLT2 inhibitors can induce erythrocytosis without iron supplementation suggests that the abnormalities in iron diagnostic tests in patients with mild-to-moderate heart failure are likely to be functional, rather than absolute, that is, they are related to inflammation-mediated trapping of iron by hepcidin and ferritin, which is reversed by treatment with SGLT2 inhibitors. An increase in bioreactive cytosolic Fe2+ is also likely to augment mitochondrial production of ATP in cardiomyocytes, thus retarding the progression of heart failure. These effects on iron metabolism are consistent with (i) proteomics analyses of placebo-controlled trials, which have shown that biomarkers of iron homeostasis represent the most consistent effect of SGLT2 inhibitors; and (ii) statistical mediation analyses, which have reported striking parallelism of the effect of SGLT2 inhibitors to promote erythrocytosis and reduce heart failure events.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular InstituteDallasTXUSA
- Imperial CollegeLondonUK
| |
Collapse
|
106
|
Del Pinto R, Ferri C. Iron deficiency in heart failure: diagnosis and clinical implications. Eur Heart J Suppl 2022; 24:I96-I99. [PMID: 36380788 PMCID: PMC9653155 DOI: 10.1093/eurheartjsupp/suac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Iron deficiency is a widely prevalent finding in patients with heart failure, observed on average in 50% of outpatients and up to 80% of acute patients, regardless of the ejection fraction and the presence of anaemia, being an independent predictor of worst functional capacity and reduced survival. The definition of iron deficiency in heart failure considers the state of chronic inflammation that characterizes the pathology, recognizing a discriminating role for transferrin saturation. The studies conducted so far, which focused on the patient with heart failure with at least moderately reduced ejection fraction, have shown clinical benefit with intravenous supplementation of ferric carboxymaltose in terms of functional capacity, quality of life, laboratory markers of disease and inflammation, and possible reduction of re-hospitalizations, but not in terms of mortality. Based on this evidence, guidelines recommend intravenous ferric carboxymaltose in decompensated and iron-deficient patients, while research is at work to investigate the clinical impact of supplementation in contexts not yet examined, such as that of decompensation in patients with heart failure and preserved ejection fraction.
Collapse
Affiliation(s)
- Rita Del Pinto
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences, University of L’Aquila
| | - Claudio Ferri
- UOC Internal Medicine and Nephrology and ESH Center of Excellence for Arterial Hypertension and Cardiovascular Prevention—S. Salvatore Regional Hospital , L’Aquila
| |
Collapse
|
107
|
López-Vilella R, Donoso Trenado V, Jover Pastor P, Sánchez-Lázaro I, Martínez Dolz L, Almenar Bonet L. Why Iron Deficiency in Acute Heart Failure Should Be Treated: A Real-World Clinical Practice Study. Life (Basel) 2022; 12:1828. [PMID: 36362983 PMCID: PMC9699465 DOI: 10.3390/life12111828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022] Open
Abstract
Background. This study aims to determine whether the administration of ferric carboxymaltose (FCM) in patients with acute heart failure (AHF) and iron deficiency (ID) improves morbidity and mortality. Methods. We studied 890 consecutive patients admitted for AHF. Patients were divided into six groups according to reduced left ventricular ejection fraction (HFrEF) or preserved (HFpEF), presence of ID, and administration of FCM. Emergency visits, re-admissions, and all-cause mortality were assessed at 6 months. Results. The overall prevalence of ID was 91.2%. In the HFrEF group, no differences were found in isolated events when patients with untreated vs. treated ID were compared, while differences were found in the combined event rate (p = 0.049). The risk calculation showed an absolute risk reduction (ARR) of 10% and relative risk reduction (RRR) of 18%. In HFpEF there was a positive trend with regard to the combined event (p = 0.107), with an ARR of 9% and an RRR of 15%. The number of patients we needed to treat to prevent a combined event was 10.5 in HFrEF and 10.8 in HFpEF. Conclusions. FCM in AHF reduced the combined event rate of emergency visits, re-admission, and all-cause death at 6 months in HF with left ventricular ejection fraction <50%, and showed a positive trend in HFpEF.
Collapse
Affiliation(s)
- Raquel López-Vilella
- Heart Failure and Transplantation Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Cardiology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Víctor Donoso Trenado
- Heart Failure and Transplantation Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Cardiology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Pablo Jover Pastor
- Department of Cardiology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Ignacio Sánchez-Lázaro
- Heart Failure and Transplantation Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Cardiology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis Martínez Dolz
- Department of Cardiology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis Almenar Bonet
- Heart Failure and Transplantation Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Cardiology, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
108
|
Hwang NC, Sivathasan C. Preoperative Evaluation and Care of Heart Transplant Candidates. J Cardiothorac Vasc Anesth 2022; 36:4161-4172. [PMID: 36028377 DOI: 10.1053/j.jvca.2022.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 06/20/2022] [Accepted: 07/08/2022] [Indexed: 11/11/2022]
Abstract
Heart transplantation is recommended for patients with advanced heart failure refractory to medical and device therapy, and who do not have absolute contraindications. When patients become eligible for heart transplantation, they undergo comprehensive evaluation and preparation to optimize their posttransplantation outcomes. This review provides an overview of the processes that are employed to enable the candidates to be transplant-ready when donor hearts are available.
Collapse
Affiliation(s)
- Nian Chih Hwang
- Department of Anaesthesiology, Singapore General Hospital, Singapore; Department of Cardiothoracic Anaesthesia, National Heart Centre, Singapore.
| | - Cumaraswamy Sivathasan
- Mechanical Cardiac Support and Heart Transplant Program, Department of Cardiothoracic Surgery, National Heart Centre, Singapore
| |
Collapse
|
109
|
Xu C, Li W, Li T, Yuan J, Pang X, Liu T, Liang B, Cheng L, Sun X, Dong S. Iron metabolism-related genes reveal predictive value of acute coronary syndrome. Front Pharmacol 2022; 13:1040845. [PMID: 36330096 PMCID: PMC9622999 DOI: 10.3389/fphar.2022.1040845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022] Open
Abstract
Iron deficiency has detrimental effects in patients with acute coronary syndrome (ACS), which is a common nutritional disorder and inflammation-related disease affects up to one-third people worldwide. However, the specific role of iron metabolism in ACS progression is opaque. In this study, we construct an iron metabolism-related genes (IMRGs) based molecular signature of ACS and to identify novel iron metabolism gene markers for early stage of ACS. The IMRGs were mainly collected from Molecular Signatures Database (mSigDB) and two relevant studies. Two blood transcriptome datasets GSE61144 and GSE60993 were used for constructing the prediction model of ACS. After differential analysis, 22 IMRGs were differentially expressed and defined as DEIGs in the training set. Then, the 22 DEIGs were trained by the Elastic Net to build the prediction model. Five genes, PADI4, HLA-DQA1, LCN2, CD7, and VNN1, were determined using multiple Elastic Net calculations and retained to obtain the optimal performance. Finally, the generated model iron metabolism-related gene signature (imSig) was assessed by the validation set GSE60993 using a series of evaluation measurements. Compared with other machine learning methods, the performance of imSig using Elastic Net was superior in the validation set. Elastic Net consistently scores the higher than Lasso and Logistic regression in the validation set in terms of ROC, PRC, Sensitivity, and Specificity. The prediction model based on iron metabolism-related genes may assist in ACS early diagnosis.
Collapse
Affiliation(s)
- Cong Xu
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Wanyang Li
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Tangzhiming Li
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Jie Yuan
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Xinli Pang
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Tao Liu
- International Digital Economy Academy, Shenzhen, China
| | - Benhui Liang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lixin Cheng
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| | - Xin Sun
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| | - Shaohong Dong
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| |
Collapse
|
110
|
Montagnani A, Frasson S, Gussoni G, Dentali F, Fontanella A, Manfellotto D. Anemia and iron in internal medicine: an Italian survey and a review on iron intravenous therapy in medical patients. ITALIAN JOURNAL OF MEDICINE 2022. [DOI: 10.4081/itjm.2022.1532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
In Italy, Internal Medicine Units hospitalize approximately 1,300,000 patients, often elderly and comorbid. The prevalent diagnoses are respiratory diseases, heart failure, or pneumonia. As a matter of fact, anemia is probably underestimated in the compilation of the official discharge forms (SDO) according to ICD-9 diagnostic codes. We promoted a survey among the Members the Italian Scientific Society of Internal Medicine (FADOI) with the aim to investigate the prevalence of anemia and iron deficiency, over than certain aspects related to the therapeutic management of patients with anemia. Furthermore, we performed a review summarizing current evidence for iron intravenous therapy in these patients. According to the survey, anemia is present in around half of the patients hospitalized in Internal Medicine, and about a quarter of them shows iron metabolism alterations. In the evaluation of iron metabolism, the dosage of ferritin is the most requested exam, whereas transferrin saturation is less considered. By focusing on some categories of patients, the awareness of the usefulness of intravenous iron therapy in patients with heart failure seems to be sufficiently common (76% of physicians), while it seems lower (60%) in the management of patients with chronic kidney disease (CKD) and anemia. Finally, more than 75% of the physicians answered that, in their hospital, there are few outpatients’ offices or diagnostic pathways dedicated to patients with anemia. Anemia due to absolute or functional iron deficiency is particularly prevalent in Internal Medicine inpatients. For this reason, an accurate evaluation of iron profile and an adequate iron therapy is mandatory in these patients. Recent studies show that, in patients with heart failure, intravenous iron therapy is an effective way of improving patients’ health, regardless of the presence of anemia. Similarly, iron therapy results fundamental to optimize erythropoiesis-stimulating agent efficacy in patients with chronic renal failure. In the next future, other therapeutic aspects of intravenous iron therapy will be probably clarified by several interesting ongoing studies focused on these patients.
Collapse
|
111
|
Song Z, Tang M, Tang G, Fu G, Ou D, Yao F, Hou X, Zhang D. Oral iron supplementation in patients with heart failure: a systematic review and meta-analysis. ESC Heart Fail 2022; 9:2779-2786. [PMID: 35758130 PMCID: PMC9715811 DOI: 10.1002/ehf2.14020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/19/2022] [Accepted: 06/03/2022] [Indexed: 11/07/2022] Open
Abstract
AIMS This review aimed to assess whether oral iron supplementation in a chronic heart failure (HF) population with iron deficiency (ID) or mild anaemia is safe and effective according to evidence-based medicine. METHODS We retrieved 1803 records from the PubMed, Embase, and the Cochrane Library databases from 1 January 1991 to 15 September 2021. The clinical outcome of oral iron supplementation for ID anaemia in patients with HF was the primary endpoint. The primary safety measures included adverse events and all-cause mortality, and efficacy measures included transferrin saturation (Tsat), ferritin levels, and the 6-min walk test (6MWT). The rate ratio (RR) was used to pool the efficacy measures. RESULTS Five randomized controlled trials that compared oral iron treatment for patients with the placebo group and included a combined total of 590 participants were analysed. No significant difference was found in all-cause death between oral iron treatment and placebo groups (RR = 0.77; 95% confidence intervals (CI), 0.46-1.29, Z = 0.98; P = 0.33). However, adverse events were not significantly higher in the iron treatment group (RR = 0.83; 95% CI, 0.60-1.16, Z = 1.07; P = 0.28). In addition, ferritin levels and Tsat were slightly increased after iron complex administration in patients with HF but were not statistically significant (ferritin: mean difference [MD] = 2.70, 95% CI, -2.41 to 7.81, Z = 1.04; P = 0.30; Tsat: MD = 27.42, 95% CI, -4.93 to 59.78, Z = 1.66; P = 0.10). No significant difference was found in exercise capacity, as indicated by the 6MWT results (MD = 59.60, 95% CI, -17.89 to 137.08, Z = 1.51; P = 0.13). We also analysed two non-randomized controlled trials with follow-up results showing that oral iron supplementation increased serum iron levels (MD = 28.87, 95% CI, 1.62-56.12, Z = 2.08; P = 0.04). CONCLUSIONS Based on the current findings, oral iron supplementation can increase serum iron levels in patients with HF and ID or mild anaemia but does not improve Tsat and 6MWT. In addition, oral iron supplementation is relatively safe.
Collapse
Affiliation(s)
- Zhiping Song
- Department of Cardiovascular MedicineYuechi County People's HospitalGuang'anChina
| | - Mingyang Tang
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Gang Tang
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Guoqi Fu
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Dengke Ou
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Fengyou Yao
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Xingzhi Hou
- Department of Cardiovascular MedicineYuechi County People's HospitalGuang'anChina
| | - Denghong Zhang
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| |
Collapse
|
112
|
Singh B, Bajaj N, Singh P, Kumar Ghosh A, Anathakrishnan R, Singh N. Iron deficiency in patients of heart failure with reduced ejection fraction. Med J Armed Forces India 2022; 78:463-468. [PMID: 36267507 PMCID: PMC9577345 DOI: 10.1016/j.mjafi.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
Background Heart Failure with reduced Ejection Fraction (HFrEF) is a common disorder affecting a large population. Iron deficiency (ID) with and without anaemia is an important variable which is often underreported and under treated in clinical practice, which contributes to patient symptoms. The present study was undertaken to study the prevalence and Spectrum of Iron Deficiency in patients of HFrEF. Methods This is a single-centre observational study. All patients with a clinical diagnosis of HFrEF presenting to the hospital were studied. Ejection Fraction (EF) was assessed on Echo and ID was diagnosed on basis of serum ferritin <100 micro g/dl or serum ferritin 100-300 micro g/dl with low Transferrin Saturation (TSAT) (< 20%). Results We have studied a total of 204 patients with a predominantly male population (73%) and a mean age of 62.88 years. Most of our patients were in mid-level functional class (mean 2.48 ± 0.50) and had low EF (mean 29.56 ± 6.52). Out of 204 patients, 88.7% patients had ID with 83% patients having absolute ID. Of the total patients with HF, 70% had anaemia. Amongst those with anaemia 93% had ID, and even without anaemia, 68% had absolute or functional ID, underlying the importance of evaluating iron status in all patients of HF irrespective of their haemoglobin levels. Conclusion This study highlights the burden of iron deficiency in heart failure patients in the Indian population and opens the way for large scale studies for better characterization of iron deficiency as well as therapeutic trials in the management of heart failure patients.
Collapse
Affiliation(s)
- Balbir Singh
- Classified Specialist (Medicine & Cardiology), 7 Air Force Hospital, Kanpur, India
| | - Nitin Bajaj
- Senior Advisor (Medicine) & Cardiologist, Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Priyanka Singh
- Senior Resident (Cardiology), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Arijit Kumar Ghosh
- Senior Advisor (Medicine) & Cardiologist, 151 Base Hospital, C/o 99 APO, India
| | - R. Anathakrishnan
- Senior Advisor (Medicine) & Cardiologist, INHS Asvini, Colaba, Mumbai, India
| | - Navreet Singh
- Senior Advisor (Medicine) & Cardiologist, Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| |
Collapse
|
113
|
Muacevic A, Adler JR. Iron Deficiency in Heart Failure: What Do We Know So Far? Cureus 2022; 14:e30348. [PMID: 36407262 PMCID: PMC9664420 DOI: 10.7759/cureus.30348] [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: 10/12/2022] [Indexed: 01/25/2023] Open
Abstract
Iron is vital for multiple biological processes in the human body. Heart failure (HF) patients are at a high risk of becoming iron deficient. Iron deficiency is a marker of severe HF and an ominous sign of poor outcomes. Iron deficiency can be absolute (low iron stores) or functional (improper functioning in the metabolic processes). The European Society of Cardiology recommends routine screening of iron stores in HF patients using ferritin and transferrin saturation. It advises iron replacement in deficient patients irrespective of the presence of anemia. Iron replacement improved HF symptoms, exercise capacity, and quality of life in deficient patients. It alleviates their disordered breathing during sleep. Therefore, the treatment of iron deficiency is an important target in managing HF. Oral iron is not effective in repleting iron stores in HF patients. Intravenous iron is an effective way to replenish iron stores in this cohort.
Collapse
|
114
|
Myint PT, Nandar PP, Thet AM, Orasanu G. Cost-effective heart failure management: Meta-analysis of IV iron therapy in iron-deficient heart failure patients. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 22:100204. [PMID: 38558909 PMCID: PMC10978398 DOI: 10.1016/j.ahjo.2022.100204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/25/2022] [Accepted: 09/06/2022] [Indexed: 04/04/2024]
Abstract
Background Iron deficiency is an important co-morbidity in heart failure patients. IV iron may improve quality of life and reduce heart failure hospitalizations, but the results of the clinical trials are varied. Objective The purpose of this meta-analysis is to assess not only the effect of IV iron in iron-deficient heart failure patients but also the quality of evidence. Methods PubMed and Cochrane databases were searched from inception to Oct 2021. Randomized clinical trials in iron-deficient, heart failure patients assessing the effect of IV iron versus placebo and with at least 12 weeks of follow-up were included. The outcomes were pooled and analyzed using a random-effect model. The quality of evidence was assessed using the GRADE approach. Results Seven studies were included in our meta-analysis. IV iron was associated with a 13.8 % decreased risk of HF hospitalizations (OR 0.59; 0.35-0.98, p = 0.040, GRADE = Low). All-cause mortality and CV mortality were not different between IV iron and placebo. But a composite outcome of HF hospitalizations or CV mortality was 17.5 % lower with IV iron (OR 0.51;0.31-0.84, p = 0.008, GRADE = Moderate). Conclusions Among heart failure patients with iron deficiency, IV iron is associated with lower HF hospitalizations. It is a relatively inexpensive regimen that can potentially improve quality of life and decrease healthcare expenditure.
Collapse
Affiliation(s)
- Phyo Thazin Myint
- Department of Hospital Medicine, Baystate Medical Center, Springfield, MA, USA
| | | | - Aye M. Thet
- Department of Hematology & Oncology, Ascension Macomb-Oakland Hospital, Warren, MI, USA
| | | |
Collapse
|
115
|
Cacoub P, Choukroun G, Cohen-Solal A, Luporsi E, Peyrin-Biroulet L, Peoc'h K, Andrieu V, Lasocki S, Puy H, Trochu JN. Iron deficiency screening is a key issue in chronic inflammatory diseases: A call to action. J Intern Med 2022; 292:542-556. [PMID: 35466452 PMCID: PMC9544998 DOI: 10.1111/joim.13503] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Iron deficiency is frequent in patients with chronic inflammatory conditions (e.g., chronic heart failure, chronic kidney disease, cancers, and bowel inflammatory diseases). Indeed, high concentrations of inflammatory cytokines increase hepcidin concentrations that lead to the sequestration of iron in cells of the reticuloendothelial system (functional iron deficiency). Iron parameters are often assessed only in the context of anemia, but iron deficiency, even without anemia, is present in about half of patients with inflammatory conditions. Iron deficiency worsens underlying chronic diseases and is an independent factor of morbidity and mortality. In daily practice, the most effective biomarkers of iron status are serum ferritin, which reflects iron storage, and transferrin saturation, which reflects the transport of iron. Serum ferritin is increased in an inflammatory context, and there is still no consensus on the threshold to be used in chronic inflammatory conditions. Nevertheless, recent recommendations of international guidelines agreed to define iron deficiency by serum ferritin <100 µg/L and/or transferrin saturation <20%. Iron parameters remain, however, insufficiently assessed in patients with chronic inflammatory conditions. Indeed, clinical symptoms of iron deficiency, such as fatigue, are not specific and often confused with those of the primary disease. Iron repletion, preferably by the intravenous route to bypass tissue sequestration, improves clinical signs and quality of life. Because of the negative impact of iron deficiency on chronic inflammatory diseases and the efficacy of intravenous iron repletion, screening of iron parameters should be part of the routine examination of all patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Patrice Cacoub
- Department of Internal Medicine and Clinical Immunology, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, Paris, France.,UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Sorbonne Universités, Paris, France.,Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Gabriel Choukroun
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, Amiens, France.,Division of Nephrology, Amiens University Hospital, Amiens, France
| | - Alain Cohen-Solal
- Cardiology Department, Lariboisière Hospital, AP-HP, Paris, France.,Université de Paris, Paris, France
| | | | - Laurent Peyrin-Biroulet
- Department of Gastroenterology and Inserm NGERE U1256, University Hospital of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - Katell Peoc'h
- APHP, Department of Clinical Biochemistry, Beaujon Hospital, Clichy, France.,Centre de Recherche sur l'Inflammation (CRI), INSERM UMR 1149, Université de Paris, Paris, France
| | - Valérie Andrieu
- APHP, Department of Hematology, Bichat-Claude Bernard Hospital, Paris, France
| | - Sigismond Lasocki
- Département d'Anesthésie Réanimation, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Hervé Puy
- Centre de Recherche Biomedicale Bichat-Beaujon, Universite Paris Diderot, Paris, France
| | - Jean-Noël Trochu
- Inserm, Institut du Thorax, CNRS, CHU de Nantes, Université Nantes, Nantes, France
| |
Collapse
|
116
|
Sindone A, Doehner W, Comin‐Colet J. Systematic review and meta-analysis of intravenous iron-carbohydrate complexes in HFrEF patients with iron deficiency. ESC Heart Fail 2022; 10:44-56. [PMID: 36178088 PMCID: PMC9871661 DOI: 10.1002/ehf2.14177] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
Iron deficiency (ID) is a common co-morbidity in patients with heart failure (HF). The present meta-analysis evaluates the effect of intravenous (IV) iron-carbohydrate complex supplementation in patients with HF with reduced ejection fraction (HFrEF) and ID/iron deficiency anaemia (IDA). Randomized controlled trials (RCTs) comparing IV iron-carbohydrate complexes with placebo/standard of care in patients with HFrEF with ID/IDA were identified using Embase (from 1957) and PubMed (from 1989) databases through 25 May 2021. Twelve RCTs including 2381 patients were included in this analysis. The majority (90.8%) of patients receiving IV iron-carbohydrate therapy were administered ferric carboxymaltose (FCM); 7.5% received iron sucrose and 1.6% received iron isomaltoside. IV iron-carbohydrate therapy significantly reduced hospitalization for worsening HF [0.53 (0.42-0.65); P < 0.0001] and first hospitalization for worsening HF or death [0.75 (0.59-0.95); P = 0.016], but did not significantly impact all-cause mortality, compared with control. IV iron-carbohydrate therapy significantly improved functional and exercise capacity compared with the control. There was no significant difference in outcome between IV iron-carbohydrate formulations when similar endpoints were measured. No significant difference in adverse events (AE) was observed between the treatment groups. IV iron-carbohydrate therapy resulted in improvements in a range of clinical outcomes and increased functional and exercise capacity, whereas AEs were not significantly different between IV iron-carbohydrate and placebo/standard of care arms. These findings align with the European Society of Cardiology's 2021 HF guidelines, which recommend the consideration of FCM in symptomatic patients with a left ventricular ejection fraction < 45% and ID.
Collapse
Affiliation(s)
- Andrew Sindone
- Heart Failure Unit and Department of Cardiac RehabilitationConcord HospitalSydneyAustralia
| | - Wolfram Doehner
- Berlin Institute of Health Center for Regenerative Therapies (BCRT) and Department of Cardiology (Virchow Klinikum)German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Josep Comin‐Colet
- Cardiology DepartmentBellvitge University Hospital, IDIBELL, University of Barcelona, L'Hospitalet de LlobregatBarcelonaSpain
| |
Collapse
|
117
|
Tan N, Cai Y, Liu J, Wang X, Ma L, Ling G, Jiang J, Wang Q, Wang Y. Effects and Safety of Oral Iron for Heart Failure with Iron Deficiency: A Systematic Review and Meta-Analysis with Trial Sequential Analysis. Cardiovasc Ther 2022; 2022:6442122. [PMID: 36186487 PMCID: PMC9509286 DOI: 10.1155/2022/6442122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/03/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background Oral iron supplement is commonly prescribed to heart failure patients with iron deficiency. However, the effects of oral iron for heart failure remain controversial. This study included randomized controlled trials (RCTs) for meta-analysis to evaluate the effects of oral iron for heart failure patients. Methods Nine databases (The Cochrane Library, Embase, PubMed, CINAHL, Web of science, CNKI, SinoMed, VIP, and Wanfang) were searched for RCTs of oral iron for heart failure from inception to October 2021. The effects were assessed with a meta-analysis using Revman 5.3 software. The trial sequential analysis was performed by TSA 0.9.5.10 beta software. The risk of bias of trials was evaluated via Risk of Bias tool. The evidence quality was assessed through GRADE tool. Results Four studies including 582 patients with heart failure and iron deficiency were enrolled. The results indicated that oral iron treatment could improve left ventricular ejection fraction (LVEF, MD = 1.52%, 95% CI: 0.69 to 2.36, P = 0.0003) and serum ferritin (MD = 1.64, 95% CI: 0.26 to 3.02, P = 0.02). However, there was no between-group difference in the 6-minute walk distances (6MWT), N terminal pro B type natriuretic peptide (NT-proBNP) or hemoglobin level when compared with control group. Subgroup analyses revealed that the effects of oral iron on 6 MWT and serum ferritin could not be affected by duration and frequency of oral iron uptakes. In trial sequential analysis of LVEF and serum ferritin, the Z-curves crossed the traditional boundary and trail sequential monitoring boundary but did not reach the required information size. Conclusion This analysis showed that oral iron could improve cardiac function measured by LVEF, and iron stores measured serum ferritin, but lack of effect on exercise capacity measured by 6 MWT, and iron stores measured by hemoglobin. Given the overall poor methodological quality and evidence quality, these findings should be treated cautiously.
Collapse
Affiliation(s)
- Nannan Tan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, China
| | - Yiqing Cai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junjie Liu
- Department of Cardiology, Nanjing Pukou Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Xiaoping Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, China
| | - Lin Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, China
| | - Guanjing Ling
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, China
| | - Jinchi Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, China
| | - Qiyan Wang
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, China
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, China
| |
Collapse
|
118
|
Fatigue in Heart Failure. J Cardiovasc Nurs 2022. [DOI: 10.1097/jcn.0000000000000940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
119
|
Persistent Iron Deficiency and Dysregulated Hepcidin Levels After Durable Left Ventricular Assist Device Therapy. ASAIO J 2022; 69:e152-e154. [PMID: 36084293 DOI: 10.1097/mat.0000000000001811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
120
|
Bardan S, Kvaslerud AB, Andresen K, Kløve SF, Edvardsen T, Gullestad L, Broch K. Intravenous ferric derisomaltose in iron-deficient patients undergoing transcatheter aortic valve implantation due to severe aortic stenosis: study protocol of the randomised controlled IIISAS trial. BMJ Open 2022; 12:e059546. [PMID: 36691165 PMCID: PMC9442485 DOI: 10.1136/bmjopen-2021-059546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 07/13/2022] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION Iron deficiency is a prevalent comorbidity in patients with severe aortic stenosis and may be associated with procedural and clinical outcomes after transcatheter aortic valve implantation (TAVI). In the Intravenous Iron Supplement for Iron Deficiency in Patients with Severe Aortic Stenosis (IIISAS) trial, we aim to examine whether a single administration of ferric derisomaltose can improve physical capacity after TAVI. METHODS AND ANALYSIS This randomised, double-blind, placebo-controlled trial aims to enrol 150 patients with iron deficiency who are scheduled for TAVI due to severe aortic stenosis. The study drug and matching placebo are administered approximately 3 months prior to TAVI, and the patients are followed for 3 months after TAVI. Inclusion criteria are iron deficiency, defined as serum ferritin<100 µg/L or ferritin between 100 and 300 µg/L in combination with a transferrin saturation<20% and written informed consent. Exclusion criteria include haemoglobin<10 g/dL, red blood cell disorders, end-stage kidney failure, intolerance to ferric derisomaltose, and ongoing infections. The primary endpoint is the baseline-adjusted distance walked on a 6 min walk test (6MWT) 3 months after TAVI. Secondary end points include quality of life, New York Heart Association functional class (NYHA functional class), and skeletal muscle strength. ETHICS AND DISSEMINATION Ethical approval was obtained from the Regional Committee for Medical and Health Research of South-Eastern Norway and The Norwegian Medicines Agency. Enrolment has begun, and results are expected in 2022. The results of the IIISAS trial will be disseminated by presentations at international and national conferences and by publications in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT04206228.
Collapse
Affiliation(s)
- Sara Bardan
- Faculty of Medicine, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anette Borger Kvaslerud
- Faculty of Medicine, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kristoffer Andresen
- Faculty of Medicine, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Sophie Foss Kløve
- Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Thor Edvardsen
- Faculty of Medicine, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Lars Gullestad
- Faculty of Medicine, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
121
|
Iso T, Matsue Y, Mizukami A, Tokano T, Isoda K, Suwa S, Miyauchi K, Yanagisawa N, Okumura Y, Minamino T. Daprodustat for anaemia in patients with heart failure and chronic kidney disease: A randomized controlled study. ESC Heart Fail 2022; 9:4291-4297. [PMID: 35983622 PMCID: PMC9773652 DOI: 10.1002/ehf2.14109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/19/2022] [Accepted: 08/04/2022] [Indexed: 01/19/2023] Open
Abstract
AIMS Hypoxia-inducible factor-prolyl hydroxylase (HIF-PH) inhibitors have been developed for the treatment of renal anaemia; however, no study has evaluated the safety and efficacy of HIF-PH inhibitors in patients with heart failure (HF). This study was designed to evaluate the safety and efficacy of daprodustat, a HIF-PH inhibitor, in patients with HF and renal anaemia. METHODS AND RESULTS We designed a pilot, multi-centre, open-label, randomized controlled study, in which 50 patients with HF complicated with chronic kidney disease and anaemia will be randomized 1:1 to either the daprodustat or control group at seven sites in Japan. Study entry requires New York Heart Association Class II HF symptoms or a history of hospitalization due to HF, an estimated glomerular filtration rate of <60 mL/min/1.73 m2 , and a haemoglobin level of 7.5 to <11.0 g/dl. Patients randomized to the daprodustat group will be treated with oral daprodustat, and the dose will be uptitrated according to the changes in the haemoglobin level from previous visits. In this study, we will evaluate the impact of HIF-PH inhibitors on cardiac function using advanced cardiovascular imaging modalities, including cardiac magnetic resonance imaging. The primary outcome is the haemoglobin level at 16 weeks of randomization, and all adverse events will be recorded and evaluated for any association with daprodustat treatment. CONCLUSION Considering the hypothetical upside and downside of using HIF-PH inhibitors in anaemic patients with HF and chronic kidney disease, and because there are virtually no safe and effective treatments for patients with anaemia not caused by iron deficiency, our study results will contribute significantly to this field.
Collapse
Affiliation(s)
- Takashi Iso
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
| | - Yuya Matsue
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
| | - Akira Mizukami
- Department of CardiologyKameda Medical CenterKamogawaJapan
| | - Takashi Tokano
- Department of CardiologyJuntendo University Urayasu HospitalUrayasuJapan
| | - Kikuo Isoda
- Department of CardiologyJuntendo University Nerima HospitalTokyoJapan
| | - Satoru Suwa
- Department of Acute Critical Care Medicine, Shizuoka HospitalJuntendo UniversityTokyoJapan
| | - Katsumi Miyauchi
- Department of Cardiovascular MedicineJuntendo Tokyo Koto Geriatric Medical CenterTokyoJapan
| | | | - Yasuo Okumura
- Division of Cardiology, Department of MedicineNihon University School of MedicineTokyoJapan
| | - Tohru Minamino
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan,Japan Agency for Medical Research and Development‐Core Research for Evolutionary Medical Science and Technology (AMED‐CREST), Japan Agency for Medical Research and DevelopmentTokyoJapan
| |
Collapse
|
122
|
Kalra PR, Cleland JG, Petrie MC, Ahmed FZ, Foley PW, Kalra PA, Lang NN, Lane RE, Macdougall IC, Pellicori P, Pope MTB, Robertson M, Squire IB, Thomson EA, Ford I. Rationale and design of a randomised trial of intravenous iron in patients with heart failure. Heart 2022; 108:1979-1985. [PMID: 35948408 DOI: 10.1136/heartjnl-2022-321304] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES For patients with a reduced left ventricular ejection fraction (LVEF) heart failure with reduced ejection fraction (HFrEF) and iron deficiency, administration of intravenous iron improves symptoms, exercise capacity and may in the following 12 months, reduce hospitalisations for heart failure. The Effectiveness of Intravenous iron treatment versus standard care in patients with heart failure and iron deficiency (IRONMAN) trial evaluated whether the benefits of intravenous iron persist in the longer term and impact on morbidity and mortality. METHODS IRONMAN is a prospective, randomised, open-label, blinded endpoint (PROBE) event-driven trial. Patients aged ≥18 years with HFrEF (LVEF ≤45%) and evidence of iron deficiency (ferritin <100 µg/L and/or TSAT <20%) were enrolled if they had either a current or recent hospitalisation for heart failure or elevated plasma concentrations of a natriuretic peptide. Participants were randomised to receive, or not to receive, intravenous ferric derisomaltose in addition to guideline-recommended therapy for HFrEF. Every 4 months, intravenous iron was administered if either ferritin was <100 µg/L or, provided ferritin was ≤400 µg/L, TSAT was <25%. The primary endpoint is a composite of total hospitalisations for heart failure and cardiovascular death. Hospitalisation and deaths due to infection are safety endpoints. RESULTS Trial recruitment was completed across 70 UK hospital sites in October 2021. Participants were followed until the end of March 2022. We plan to report the results by November 2022. CONCLUSIONS IRONMAN will determine whether repeated doses of intravenous ferric derisomaltose are beneficial and safe for the long-term treatment of a broad range of patients with HFrEF and iron deficiency. TRIAL REGISTRATION NUMBER NCT02642562.
Collapse
Affiliation(s)
- Paul R Kalra
- Department of Cardiology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK.,School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - John Gf Cleland
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK.,Golden Jubilee National Hospital, Clydebank, UK
| | - Fozia Z Ahmed
- Division of Cardiovascular Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
| | | | - Philip A Kalra
- Department of Renal Medicine, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Ninian N Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Rebecca E Lane
- Part of Guy's and St Thomas' NHS Foundation Trust, Royal Brompton and Harefield Hospitals, London, UK
| | | | | | - Michael T B Pope
- Department of Cardiology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Michele Robertson
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,NIHR Leicester Biomedical Research Centre Cardiovascular Diseases, Leicester, UK
| | - Elizabeth A Thomson
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| |
Collapse
|
123
|
Clinical and Molecular Aspects of Iron Metabolism in Failing Myocytes. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081203. [PMID: 36013382 PMCID: PMC9409945 DOI: 10.3390/life12081203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) is a common disease that causes significant limitations on the organism's capacity and, in extreme cases, leads to death. Clinically, iron deficiency (ID) plays an essential role in heart failure by deteriorating the patient's condition and is a prognostic marker indicating poor clinical outcomes. Therefore, in HF patients, supplementation of iron is recommended. However, iron treatment may cause adverse effects by increasing iron-related apoptosis and the production of oxygen radicals, which may cause additional heart damage. Furthermore, many knowledge gaps exist regarding the complex interplay between iron deficiency and heart failure. Here, we describe the current, comprehensive knowledge about the role of the proteins involved in iron metabolism. We will focus on the molecular and clinical aspects of iron deficiency in HF. We believe that summarizing the new advances in the translational and clinical research regarding iron deficiency in heart failure should broaden clinicians' awareness of this comorbidity.
Collapse
|
124
|
Neglected Comorbidity of Chronic Heart Failure: Iron Deficiency. Nutrients 2022; 14:nu14153214. [PMID: 35956390 PMCID: PMC9370238 DOI: 10.3390/nu14153214] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
Iron deficiency is a significant comorbidity of heart failure (HF), defined as the inability of the myocardium to provide sufficient blood flow. However, iron deficiency remains insufficiently detected. Iron-deficiency anemia, defined as a decrease in hemoglobin caused by iron deficiency, is a late consequence of iron deficiency, and the symptoms of iron deficiency, which are not specific, are often confused with those of HF or comorbidities. HF patients with iron deficiency are often rehospitalized and present reduced survival. The correction of iron deficiency in HF patients is associated with improved functional capacity, quality of life, and rehospitalization rates. Because of the inflammation associated with chronic HF, which complicates the picture of nutritional deficiency, only the parenteral route can bypass the tissue sequestration of iron and the inhibition of intestinal iron absorption. Given the negative impact of iron deficiency on HF progression, the frequency and financial implications of rehospitalizations due to decompensation episodes, and the efficacy of this supplementation, screening for this frequent comorbidity should be part of routine testing in all HF patients. Indeed, recent European guidelines recommend screening for iron deficiency (serum ferritin and transferrin saturation coefficient) in all patients with suspected HF, regular iron parameters assessment in all patients with HF, and intravenous iron supplementation in symptomatic patients with proven deficiency. We thus aim to summarize all currently available data regarding this common and easily improvable comorbidity.
Collapse
|
125
|
Mei Z, Chen J, Luo S, Jin L, Liu Q, Chen Y. Comparative efficacy of intravenous and oral iron supplements for the treatment of iron deficiency in patients with heart failure: A network meta-analysis of randomized controlled trials. Pharmacol Res 2022; 182:106345. [PMID: 35810949 DOI: 10.1016/j.phrs.2022.106345] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVE We aimed at comparing the efficacy of intravenous and oral iron supplementations for the treatment of iron deficiency (ID) in patients with heart failure (HF). METHODS We searched the PubMed, Cochrane, and Embase databases from inception to January 15, 2022. We included randomized controlled trials enrolling patients with HF who were treated for ID with intravenous iron supplements, oral iron supplements, or placebo. The primary outcomes were all-cause death, cardiovascular mortality, and hospitalization for heart failure. The secondary outcomes were evaluated through the six-minute walking test (6MWT) and the Kansas City Cardiomyopathy Questionnaire (KCCQ). RESULTS The network meta-analysis included sixteen studies. Compared to placebo/control groups, intravenous iron supplements did not decrease all-cause death (0.69, 0.39-1.23) or cardiovascular mortality (0.89, 0.66-1.20). After 12 weeks, a reduced hospitalization for heart failure was associated with the administration of intravenous iron supplementations (0.58, 0.34-0.97). The most significant improvements regarding 6MWT (44.44, 6.10-82.79) and KCCQ (5.96, 3.19-8.73) were observed with intravenous iron supplements. Oral iron supplements reduced hospitalization for heart failure (0.36, 0.14-0.96) and all-cause death (0.34, 0.12-0.95), but did not influence the 6MWT (29.74, -47.36 to 106.83) and KCCQ (0.10, -10.95 to 11.15). CONCLUSIONS Administering intravenous iron supplements for ID in patients with HF improves their exercise capacity and quality of life. In order to reduce hospitalizations for heart failure, the supplementation should be administered for more than 12 weeks. Although oral iron supplements did not improve exercise capacity and quality of life, they could reduce all-cause death and hospitalizations for heart failure.
Collapse
Affiliation(s)
- Ziwei Mei
- Lishui Municipal Central Hospital, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, China
| | - Jun Chen
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China.
| | - Songmei Luo
- Lishui Municipal Central Hospital, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, China
| | - Lie Jin
- Lishui Municipal Central Hospital, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, China
| | - Qiang Liu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yijie Chen
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
126
|
von Haehling S. Erhalt der Selbstständigkeit bei Herzinsuffizienz: Ansatzpunkte und Konsequenzen für den Alltag. AKTUELLE KARDIOLOGIE 2022. [DOI: 10.1055/a-1820-8230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
ZusammenfassungErhalt von Mobilität und sozialer Interaktion hat für Patienten mit Herzinsuffizienz enorme Alltagsbedeutung, die in vielen bisher durchgeführten Therapiestudien nicht in ausreichendem Maße
abgebildet wurde. Ivabradin, die SGLT2-Inhibitoren Empagliflozin und Dapagliflozin sowie der ARNI Sacubitril/Valsartan bieten hier erste Möglichkeiten der Einflussnahme. Auch
Ausdauertraining ist sehr zu empfehlen. Die Therapie von Komorbiditäten bei Herzinsuffizienz zeigt vor allem bei der Therapie des Eisenmangels gute Möglichkeiten der Besserung der
Belastbarkeit, außerdem durch die Pulmonalvenenisolation bei Vorhofflimmern. Andere Aspekte, welche die Mobilität der Patienten verbessern, sind das Ermöglichen von selbstständigem Führen
von Fahrzeugen, von Sport und Hobbys, Berufstätigkeit und Sexualität sowie das Ermöglichen von Reiseaktivitäten, wenn die Patienten entsprechend vorbereitet sind, über ausreichend
Informationen für die Reiseaktivität verfügen und das Reiseziel entsprechend ausgewählt wurde. Wichtig ist, die Bedürfnisse des Patienten zu erfragen, um individualisierte Therapiekonzepte
zu erarbeiten.
Collapse
Affiliation(s)
- Stephan von Haehling
- Klinik für Kardiologie und Pneumologie, Universitätsmedizin Göttingen, Göttingen, Deutschland
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Standort Göttingen, Deutschland
| |
Collapse
|
127
|
Abstract
INTRODUCTION Iron deficiency is associated with worse outcomes in children and adults with systolic heart failure. While oral iron replacement has been shown to be ineffective in adults with heart failure, its efficacy in children with heart failure is unknown. We hypothesised that oral iron would be ineffective in replenishing iron stores in ≥50% of children with heart failure. METHODS We performed a single-centre retrospective cohort study of patients aged ≤21 years with systolic heart failure and iron deficiency who received oral iron between 01/2013 and 04/2019. Iron deficiency was defined as ≥2 of the following: serum iron <50 mcg/dL, serum ferritin <20 ng/mL, transferrin >300 ng/mL, transferrin saturation <15%. Iron studies and haematologic indices pre- and post-iron therapy were compared using paired-samples Wilcoxon test. RESULTS Fifty-one children with systolic heart failure and iron deficiency (median age 11 years, 49% female) met inclusion criteria. Heart failure aetiologies included cardiomyopathy (51%), congenital heart disease (37%), and history of heart transplantation with graft dysfunction (12%). Median dose of oral iron therapy was 2.9 mg/kg/day of elemental iron, prescribed for a median duration of 96 days. Follow-up iron testing was available for 20 patients, of whom 55% (11/20) remained iron deficient despite oral iron therapy. CONCLUSIONS This is the first report on the efficacy of oral iron therapy in children with heart failure. Over half of the children with heart failure did not respond to oral iron and remained iron deficient.
Collapse
|
128
|
Siddiqui SW, Ashok T, Patni N, Fatima M, Lamis A, Anne KK. Anemia and Heart Failure: A Narrative Review. Cureus 2022; 14:e27167. [PMID: 36017290 PMCID: PMC9393312 DOI: 10.7759/cureus.27167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2022] [Indexed: 11/06/2022] Open
Abstract
Anemia in heart failure patients is a relatively common finding and has been linked with an increased risk of hospital admissions, morbidities, and significant mortality making its correction a significant factor in improving the quality of life and clinical outcomes in those suffering from it. This review article has discussed the multifactorial pathophysiology, including iron deficiency, longstanding inflammation, abnormal levels of human erythropoietin (Epo), and the abnormal activation of the renin-angiotensin-aldosterone system (RAAS) being the most significant. The diagnostic guidelines as well as research-based management modalities specifically with iron supplements and erythropoietin stimulating agents have also been discussed, although research done in this area has been limited and shown conflicting results.
Collapse
|
129
|
Rahman EU, Chobufo MD, Farah F, Mohamed T, Elhamdani M, Rueda C, Aronow WS, Fonarow GC, Thompson E. Prevalence and temporal trends of anemia in patients with heart failure. QJM 2022; 115:437-441. [PMID: 34264349 DOI: 10.1093/qjmed/hcab193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/17/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Anemia is an important comorbidity in heart failure (HF), and it is associated with increased adverse disease experience and mortality. Previous reports have focused mainly on HF presenting in healthcare settings. We, therefore, set out to establish the nationwide prevalence and temporal trends of anemia in community-based patients with HF in the US. AIM To establish the nationwide prevalence and temporal trends of anemia in community-based patients with HF in the US. DESIGN The NHANES dataset, conducted by the CDC National Center for Health Statistics was used to collect nationally representative data on the health and nutritional status of the non-institutionalized US population. METHODS We utilized the National Health and Nutrition Examination data collected over five survey cycles (2007-16). Included were participants aged 20-80 years with self-reported diagnosis of HF. Anemia was defined using 2 sex specific cut offs of 13 and 12 g/dl (cutoff 1), and 12 and 11 g/dl (cutoff 2), for men and women, respectively. The Chi square test was used to compare prevalence across different categories and survey cycles. Data analysis was done using STATA 16 with P-values < 0.05 considered statistically significant. RESULTS The median hemoglobin in all HF patients was 13.5 g/dl (IQR 12.4-14.5). The prevalence of anemia among community-based patients with HF in the US was 21.34% (cutoff 1) and 9.03% (cutoff 2) and has been stable from 2007 to 2016. The burden of anemia was disproportionately higher in NH Blacks (34.48%, 95% CI 27.12-42.67) and those with BMI < 25 Kg/m2 (17.4%, 95% CI 10.9-26.64). CONCLUSION The prevalence of anemia in patients with HF in the US is at least 9% and has remained stable over the past decade. This high persistent burden with limited proven interventions should spur further efforts aimed at identifying impactful ways of addressing anemia in patients with HF.
Collapse
Affiliation(s)
- E U Rahman
- From the Department of Internal Medicine, St Mary's Medical Center, 2900 First Avenue, Huntington, WV 25702, USA
| | - M D Chobufo
- Department of Internal Medicine, Interfaith Medical Center, 1545 Atlantic Ave, Brooklyn, NY 11213, USA
| | - F Farah
- Department of Internal Medicine, Deccan College of Medical Sciences, DMRL X Road, Santosh Nagar Main Rd, Kanchan Bagh, Hyderabad, Telangana 500058, India
| | - T Mohamed
- Department of Cardiology, Marshall University, 1 John Marshall dr, Huntington, WV 25755, USA
| | - M Elhamdani
- Department of Cardiology, Marshall University, 1 John Marshall dr, Huntington, WV 25755, USA
| | - C Rueda
- Department of Cardiology, Marshall University, 1 John Marshall dr, Huntington, WV 25755, USA
| | - W S Aronow
- Department of Cardiology research, Westchester Medical Centre and New York Medical College, 100 Woods Rd, Valhalla, NY 10595, USA
| | - G C Fonarow
- Department of Cardiology, Ronald Reagan-UCLA Medical Center, 757 Westwood Plaza, Los Angeles, CA 90095, USA
| | - E Thompson
- Department of Cardiology, Marshall University, 1 John Marshall dr, Huntington, WV 25755, USA
| |
Collapse
|
130
|
Cuthbert JJ, Ransome N, Clark AL. Re-defining iron deficiency in patients with heart failure. Expert Rev Cardiovasc Ther 2022; 20:667-681. [DOI: 10.1080/14779072.2022.2100349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- JJ Cuthbert
- Department of Cardiorespiratory Medicine, Centre for Clinical Sciences, Hull York Medical School, University of Hull, Kingston-Upon-Hull, East Riding of Yorkshire, UK
- Department of Cardiology, Hull University Teaching Hospital Trust, Castle Hill Hospital, Castle Road, Cottingham, Kingston-Upon-Hull, East Riding of Yorkshire, UK
| | - N Ransome
- Department of Haematology, York and Scarborough Teaching Hospitals NHS Trust, York, UK
| | - AL Clark
- Department of Cardiology, Hull University Teaching Hospital Trust, Castle Hill Hospital, Castle Road, Cottingham, Kingston-Upon-Hull, East Riding of Yorkshire, UK
| |
Collapse
|
131
|
Essa H, Walker L, Mohee K, Oguguo C, Douglas H, Kahn M, Rao A, Bellieu J, Hadcroft J, Hartshorne-Evans N, Bliss J, Akpan A, Wong C, Cuthbertson DJ, Sankaranarayanan R. Multispecialty multidisciplinary input into comorbidities along with treatment optimisation in heart failure reduces hospitalisation and clinic attendance. Open Heart 2022; 9:openhrt-2022-001979. [PMID: 35858706 PMCID: PMC9305818 DOI: 10.1136/openhrt-2022-001979] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Aims Heart failure (HF) is associated with comorbidities which independently influence treatment response and outcomes. This retrospective observational study (January 2020–June 2021) analysed the impact of monthly HF multispecialty multidisciplinary team (MDT) meetings to address management of HF comorbidities and thereby on provision, cost of care and HF outcomes. Methods Patients acted as their own controls, with outcomes compared for equal periods (for each patient) pre (HF MDT) versus post-MDT (multispecialty) meeting. The multispecialty MDT comprised HF cardiologists (primary, secondary, tertiary care), HF nurses, nephrologist, endocrinologist, palliative care, chest physician, pharmacist, clinical pharmacologist and geriatrician. Outcome measures were (1) all-cause hospitalisations, (2) outpatient clinic attendances and (3) cost. Results 334 patients (mean age 72.5±11 years) were discussed virtually through MDT meetings and follow-up duration was 13.9±4 months. Mean age-adjusted Charlson Comorbidity Index was 7.6±2.1 and Rockwood Frailty Score 5.5±1.6. Multispecialty interventions included optimising diabetes therapy (haemoglobin A1c-HbA1c pre-MDT 68±11 mmol/mol vs post-MDT 61±9 mmol/mol; p<0.001), deprescribing to reduce anticholinergic burden (pre-MDT 1.85±0.4 vs 1.5±0.3 post-MDT; p<0.001), initiation of renin–angiotensin aldosterone system inhibitors in HF with reduced ejection fraction (HFrEF) with advanced chronic kidney disease (9% pre vs 71% post-MDT; p<0.001). Other interventions included potassium binders, treatment of anaemia, falls assessment, management of chest conditions, day-case ascitic, pleural drains and palliative support. Total cost of funding monthly multispecialty meetings was £32 400 and resultant 64 clinic appointments cost £9600. The post-MDT study period was associated with reduction in 481 clinic appointments (cost saving £72150) and reduced all-cause hospitalisations (pre-MDT 1.1±0.4 vs 0.6±0.1 post-MDT; p<0.001), reduction of 1586 hospital bed-days and cost savings of £634 400. Total cost saving to the healthcare system was £664 550. Conclusion HF multispecialty virtual MDT model provides integrated, holistic care across all healthcare tiers for management of HF and associated comorbidities. This approach is associated with reduced clinic attendances and all-cause hospitalisations, leading to significant cost savings.
Collapse
Affiliation(s)
- Hani Essa
- Cardiology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.,Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| | - Lauren Walker
- Pharmacology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.,Clinical Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Kevin Mohee
- Cardiology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Chukwuemeka Oguguo
- Cardiology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Homeyra Douglas
- Cardiology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Matthew Kahn
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK.,Cardiology, Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Archana Rao
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK.,Cardiology, Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Julie Bellieu
- Palliative Medicine, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Justine Hadcroft
- Respiratory Medicine, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Nick Hartshorne-Evans
- CEO and Founder, The Pumping Marvellous Foundation (Patient-Led Heart Failure Charity), Preston, UK
| | - Janet Bliss
- Chair, NHS Liverpool Clinical Commissioning Group, Liverpool, UK.,GP Senior Partner, Grey Road Surgery, Liverpool, UK
| | - Asangaedem Akpan
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK.,Institute of Health, University of Cumbria, Cumbria, UK.,Geriatrics, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.,Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Christopher Wong
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK.,Nephrology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.,School of Health and Sport Sciences, Liverpool Hope University, Liverpool, UK
| | - Daniel J Cuthbertson
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK.,Faculty of Health and Life Science, University of Liverpool, Liverpool, UK.,Diabetes and Endocrinology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Rajiv Sankaranarayanan
- Cardiology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK .,Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK.,NIHR Research Scholar, NIHR CRN North West Coast, National Institute for Health and Care Research (NIHR), Liverpool, UK
| |
Collapse
|
132
|
VENTURINI E, IANNUZZO G, DI LORENZO A, CUOMO G, D'ANGELO A, MERONE P, CUDEMO G, PACILEO M, D'ANDREA A, VIGORITO C, GIALLAURIA F. Short-term treatment of iron deficiency anemia after cardiac surgery. IJC HEART & VASCULATURE 2022; 40:101038. [PMID: 35514874 PMCID: PMC9066354 DOI: 10.1016/j.ijcha.2022.101038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022]
Abstract
Iron deficiency anemia (IDA) is frequent after cardiac surgery and is associated with increased morbidity and mortality. In a retrospective study, we analyzed 106 patients with IDA (hemoglobin [Hb] ≤ 12 g/dl in women and ≤ 13 g/dl in men, transferrin saturation [TSAT] ≤ 20%) on admission to a Cardiac Rehabilitation Unit after cardiac surgery. The patients were divided into two groups, one was treated with oral sucrosomial iron (SI) and the other with intravenous ferric carboxymaltose (FCM). Patients received a single 1000 mg dose of FCM from the day after admission to rehabilitation (T1), or a 120 mg/day dose of SI from T1 until discharge (T2); after discharge, SI was reduced to 30 mg/day until the end of follow-up (T3). Hb was evaluated at T1, T2 and T3; the other hematological parameters at T1 and T3; natriuretic peptides at T1, T2 and T3; 6-minute walk test (6MWT) at T1 and T2. Folate, vitamin B12 and reticulocytes were sampled on admission. Folate deficiency was documented in 60.4% of patients. Hb increased in both groups with no significant differences between the two treatments (p = 0.397). The other iron metabolism parameters (sideremia, transferrin, TSAT) displayed similar behavior, showing a significant increase at T3 (p < 0.001) with both therapies, although the increase was faster with FCM. Ferritin - high on admission - decreased at T3 in the SI group and rose significantly in the FCM group (SI 219.5 vs. FCM 689 ng/ml p < 0.0001). The 6MWT increased significantly at T2, with an overlap between SI and FCM. In conclusion, the results of this study show that SI and FCM exhibit the same effectiveness on IDA; the response time to therapy of both treatments is also equally fast. SI and FCM induce a similar increase in functional capacity. The study shows that SI can be a viable alternative to FCM after cardiac surgery in terms of effectiveness and tolerability.
Collapse
Affiliation(s)
- Elio VENTURINI
- Cardiac Rehabilitation Unit and Department of Cardiology, Azienda USL Toscana Nord-Ovest, “Cecina Civil Hospital”, 57023 - Cecina (LI), Italy
| | - Gabriella IANNUZZO
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 – Naples, Italy
| | - Anna DI LORENZO
- Department of Translational Medical Sciences, Division of Internal Medicine and Cardiac Rehabilitation, “Federico II” University of Naples, 80131 – Naples, Italy
| | - Gianluigi CUOMO
- Department of Translational Medical Sciences, Division of Internal Medicine and Cardiac Rehabilitation, “Federico II” University of Naples, 80131 – Naples, Italy
| | - Andrea D'ANGELO
- Department of Translational Medical Sciences, Division of Internal Medicine and Cardiac Rehabilitation, “Federico II” University of Naples, 80131 – Naples, Italy
| | - Pasquale MERONE
- Department of Translational Medical Sciences, Division of Internal Medicine and Cardiac Rehabilitation, “Federico II” University of Naples, 80131 – Naples, Italy
| | - Giuseppe CUDEMO
- Department of Translational Medical Sciences, Division of Internal Medicine and Cardiac Rehabilitation, “Federico II” University of Naples, 80131 – Naples, Italy
| | - Mario PACILEO
- Department of Cardiology and Intensive Coronary Care, “Umberto I Hospital”, 84014 - Nocera Inferiore (SA), Italy
| | - Antonello D'ANDREA
- Department of Cardiology and Intensive Coronary Care, “Umberto I Hospital”, 84014 - Nocera Inferiore (SA), Italy
| | - Carlo VIGORITO
- Department of Translational Medical Sciences, Division of Internal Medicine and Cardiac Rehabilitation, “Federico II” University of Naples, 80131 – Naples, Italy
| | - Francesco GIALLAURIA
- Department of Translational Medical Sciences, Division of Internal Medicine and Cardiac Rehabilitation, “Federico II” University of Naples, 80131 – Naples, Italy
| |
Collapse
|
133
|
Bomer N, Pavez-Giani MG, Grote Beverborg N, Cleland JGF, van Veldhuisen DJ, van der Meer P. Micronutrient deficiencies in heart failure: Mitochondrial dysfunction as a common pathophysiological mechanism? J Intern Med 2022; 291:713-731. [PMID: 35137472 PMCID: PMC9303299 DOI: 10.1111/joim.13456] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Heart failure is a devastating clinical syndrome, but current therapies are unable to abolish the disease burden. New strategies to treat or prevent heart failure are urgently needed. Over the past decades, a clear relationship has been established between poor cardiac performance and metabolic perturbations, including deficits in substrate uptake and utilization, reduction in mitochondrial oxidative phosphorylation and excessive reactive oxygen species production. Together, these perturbations result in progressive depletion of cardiac adenosine triphosphate (ATP) and cardiac energy deprivation. Increasing the delivery of energy substrates (e.g., fatty acids, glucose, ketones) to the mitochondria will be worthless if the mitochondria are unable to turn these energy substrates into fuel. Micronutrients (including coenzyme Q10, zinc, copper, selenium and iron) are required to efficiently convert macronutrients to ATP. However, up to 50% of patients with heart failure are deficient in one or more micronutrients in cross-sectional studies. Micronutrient deficiency has a high impact on mitochondrial energy production and should be considered an additional factor in the heart failure equation, moving our view of the failing myocardium away from an "an engine out of fuel" to "a defective engine on a path to self-destruction." This summary of evidence suggests that supplementation with micronutrients-preferably as a package rather than singly-might be a potential therapeutic strategy in the treatment of heart failure patients.
Collapse
Affiliation(s)
- Nils Bomer
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Mario G Pavez-Giani
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Niels Grote Beverborg
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - John G F Cleland
- Robertson Centre for Biostatistics and Clinical Trials, University of Glasgow, Glasgow, UK.,National Heart & Lung Institute, Royal Brompton and Harefield Hospitals, Imperial College, London, UK
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
134
|
MEHEDINTI AM, CAPUSA C, ANDREIANA I, MIRCESCU G. Intravenous Iron-Carbohydrate Nanoparticles and Their Similars. What Do We Choose? MAEDICA 2022; 17:436-448. [PMID: 36032600 PMCID: PMC9375892 DOI: 10.26574/maedica.2022.17.2.436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Anemia is highly prevalent worldwide and iron deficiency is the first cause. Iron deficiency has not only hematologic effects but also non-hematologic effects - immune, metabolic, cognitive dysfunctions and poor cardiovascular and renal outcomes - which generally precede anemia. Iron therapy not only significantly improves the hematological parameters but also has non-hematologic benefits. Given that its efficacy and safety has been revealed over the years, intravenous (IV) iron therapy is frequently used. Intravenous iron products are nanoparticles largely consisting in an iron core surrounded by a carbohydrate shell. They are classified as non-biological complex molecules, being different from small commonly used molecules, with properties and biological behavior impossible to be completely characterized only by physicochemical analysis. To date, there is no appropriate regulatory evaluation system for these medicines and several follow-on versions of the IV iron originators (e.g., iron sucrose) were approved using the same regulatory pathway as for generics. Because of this vulnerability in an adequate pathway for approval, both non-clinical and clinical studies suggested no therapeutic equivalence (thus no interchangeability) between iron sucrose originator (Venofer®), and iron sucrose similars. In this review we aimed to underline the importance of intravenous iron therapy as well as raise awareness regarding the differences between nanomedicines and their intended similar but not identical copies. The potential implications of these differences impact patients (safety, efficacy) but also the medical system (higher costs).
Collapse
Affiliation(s)
- Ana Maria MEHEDINTI
- “Carol Davila” University of Medicine and Pharmacy, Nephrology Department, Bucharest, Romania,“Dr. Carol Davila” Teaching Hospital of Nephrology, Bucharest, Romania
| | - Cristina CAPUSA
- “Carol Davila” University of Medicine and Pharmacy, Nephrology Department, Bucharest, Romania,“Dr. Carol Davila” Teaching Hospital of Nephrology, Bucharest, Romania
| | - Iuliana ANDREIANA
- “Carol Davila” University of Medicine and Pharmacy, Nephrology Department, Bucharest, Romania,“Dr. Carol Davila” Teaching Hospital of Nephrology, Bucharest, Romania
| | - Gabriel MIRCESCU
- “Carol Davila” University of Medicine and Pharmacy, Nephrology Department, Bucharest, Romania,“Dr. Carol Davila” Teaching Hospital of Nephrology, Bucharest, Romania
| |
Collapse
|
135
|
Uskach TM. Management of iron deficiency in chronic heart failure. TERAPEVT ARKH 2022; 94:572-578. [DOI: 10.26442/00403660.2022.04.201451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022]
Abstract
Iron deficiency is frequent in patients with chronic heart failure (CHF) with a prevalence of 50%, and its frequency varies depending on the study groups. The presence of iron deficiency limits erythropoiesis, leading to the development of anemia over time in patients with CHF, regardless of gender, race, and left ventricular ejection fraction (LVEF). Observational studies demonstrate a higher prevalence of iron deficiency in women and in patients with higher NYHA (New York Heart Association) functional class, decreased LVEF, increased brain natriuretic peptide (NT-proBNP), or increased high-sensitivity C-reactive protein. Iron deficiency and anemia in patients with CHF are independently associated with a decreased exercise capacity, hospitalizations for CHF, an increase in overall mortality and mortality from cardiovascular diseases. The clinical significance of iron deficiency requires the need to diagnose iron metabolism in all patients with CHF. Current guidelines for the diagnosis and treatment of CHF indicate the need to determine the level of ferritin and saturation of transferrin in all patients with a suspected diagnosis of heart failure. The use of oral iron therapy in patients with CHF demonstrates its low efficacy in correcting this condition according to the clinical trials. At the same time the use of intravenous iron therapy is safe and improves symptoms, exercise capacity and quality of life in patients with heart failure with reduced ejection fraction and iron deficiency, which has been shown both in international placebo-controlled trials and meta-analyses. The use of iron carboxymaltose should improve CHF symptoms, exercise capacity and quality of life in patients with CHF and LVEF45%. Intravenous iron therapy has also been shown to reduce readmissions for CHF in patients with an LVEF50% who have recently been hospitalized for worsening CHF.
Collapse
|
136
|
Practical Guidance for Diagnosing and Treating Iron Deficiency in Patients with Heart Failure: Why, Who and How? J Clin Med 2022; 11:jcm11112976. [PMID: 35683366 PMCID: PMC9181459 DOI: 10.3390/jcm11112976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 01/27/2023] Open
Abstract
Iron deficiency (ID) is a comorbid condition frequently seen in patients with heart failure (HF). Iron has an important role in the transport of oxygen, and is also essential for skeletal and cardiac muscle, which depend on iron for oxygen storage and cellular energy production. Thus, ID per se, even without anaemia, can be harmful. In patients with HF, ID is associated with a poorer quality of life (QoL) and exercise capacity, and a higher risk of hospitalisations and mortality, even in the absence of anaemia. Despite its negative clinical consequences, ID remains under-recognised. However, it is easily diagnosed and managed, and the recently revised 2021 European Society of Cardiology (ESC) guidelines on HF provide specific recommendations for its diagnosis and treatment. Prospective randomised controlled trials in patients with symptomatic HF with reduced ejection fraction (HFrEF) show that correction of ID using intravenous iron (principally ferric carboxymaltose [FCM]) provides improvements in symptoms of HF, exercise capacity and QoL, and a recent trial demonstrated that FCM therapy following hospitalisation due to acute decompensated HF reduced the risk of subsequent HF hospitalisations. This review provides a summary of the epidemiology and pathophysiology of ID in HFrEF, and practical guidance on screening, diagnosing, and treating ID.
Collapse
|
137
|
Roth-Walter F. Iron-Deficiency in Atopic Diseases: Innate Immune Priming by Allergens and Siderophores. FRONTIERS IN ALLERGY 2022; 3:859922. [PMID: 35769558 PMCID: PMC9234869 DOI: 10.3389/falgy.2022.859922] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Although iron is one of the most abundant elements on earth, about a third of the world's population are affected by iron deficiency. Main drivers of iron deficiency are beside the chronic lack of dietary iron, a hampered uptake machinery as a result of immune activation. Macrophages are the principal cells distributing iron in the human body with their iron restriction skewing these cells to a more pro-inflammatory state. Consequently, iron deficiency has a pronounced impact on immune cells, favoring Th2-cell survival, immunoglobulin class switching and primes mast cells for degranulation. Iron deficiency during pregnancy increases the risk of atopic diseases in children, while both children and adults with allergy are more likely to have anemia. In contrast, an improved iron status seems to protect against allergy development. Here, the most important interconnections between iron metabolism and allergies, the effect of iron deprivation on distinct immune cell types, as well as the pathophysiology in atopic diseases are summarized. Although the main focus will be humans, we also compare them with innate defense and iron sequestration strategies of microbes, given, particularly, attention to catechol-siderophores. Similarly, the defense and nutritional strategies in plants with their inducible systemic acquired resistance by salicylic acid, which further leads to synthesis of flavonoids as well as pathogenesis-related proteins, will be elaborated as both are very important for understanding the etiology of allergic diseases. Many allergens, such as lipocalins and the pathogenesis-related proteins, are able to bind iron and either deprive or supply iron to immune cells. Thus, a locally induced iron deficiency will result in immune activation and allergic sensitization. However, the same proteins such as the whey protein beta-lactoglobulin can also transport this precious micronutrient to the host immune cells (holoBLG) and hinder their activation, promoting tolerance and protecting against allergy. Since 2019, several clinical trials have also been conducted in allergic subjects using holoBLG as a food for special medical purposes, leading to a reduction in the allergic symptom burden. Supplementation with nutrient-carrying lipocalin proteins can circumvent the mucosal block and nourish selectively immune cells, therefore representing a new dietary and causative approach to compensate for functional iron deficiency in allergy sufferers.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Franziska Roth-Walter ;
| |
Collapse
|
138
|
Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022; 145:e895-e1032. [PMID: 35363499 DOI: 10.1161/cir.0000000000001063] [Citation(s) in RCA: 1099] [Impact Index Per Article: 366.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIM The "2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure" replaces the "2013 ACCF/AHA Guideline for the Management of Heart Failure" and the "2017 ACC/AHA/HFSA Focused Update of the 2013 ACCF/AHA Guideline for the Management of Heart Failure." The 2022 guideline is intended to provide patient-centric recommendations for clinicians to prevent, diagnose, and manage patients with heart failure. METHODS A comprehensive literature search was conducted from May 2020 to December 2020, encompassing studies, reviews, and other evidence conducted on human subjects that were published in English from MEDLINE (PubMed), EMBASE, the Cochrane Collaboration, the Agency for Healthcare Research and Quality, and other relevant databases. Additional relevant clinical trials and research studies, published through September 2021, were also considered. This guideline was harmonized with other American Heart Association/American College of Cardiology guidelines published through December 2021. Structure: Heart failure remains a leading cause of morbidity and mortality globally. The 2022 heart failure guideline provides recommendations based on contemporary evidence for the treatment of these patients. The recommendations present an evidence-based approach to managing patients with heart failure, with the intent to improve quality of care and align with patients' interests. Many recommendations from the earlier heart failure guidelines have been updated with new evidence, and new recommendations have been created when supported by published data. Value statements are provided for certain treatments with high-quality published economic analyses.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anita Deswal
- ACC/AHA Joint Committee on Clinical Practice Guidelines Liaison
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Neoh K, Page A, Chin-Yee N, Doree C, Bennett MI. Practice review: Evidence-based and effective management of anaemia in palliative care patients. Palliat Med 2022; 36:783-794. [PMID: 35331051 PMCID: PMC9087312 DOI: 10.1177/02692163221081967] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Anaemia is a common sequela of advanced disease and is associated with significant symptom burden. No specific guidance exists for the investigation and management of anaemia in palliative care patients. AIM We aim to offer a pragmatic overview of the approaches to investigate and manage anaemia in advanced disease, based on guidelines and evidence in disease specific patient groups, including cancer, heart failure and chronic kidney disease. DESIGN Scoping review methodology was used to determine the strength of evidence supporting the investigation and management of anaemia in patients with advanced disease. DATA SOURCES A search for guidelines was performed in 2020. National or international guidelines were examined if they described the investigation or management of anaemia in adult patients with health conditions seen by palliative care services written within the last 5 years in the English language. Searches of MEDLINE, the Cochrane library and WHO guidance were made in 2019 to identify key publications that provided additional primary data. RESULTS Evidence supports patient-centred investigation of anaemia, results of which should guide targeted intervention. Blanket use of blood transfusion should be avoided, with evidence supporting a more restrictive approach to transfusion. Routine use of oral iron and erythropoetin stimulating agents (ESAs) are not recommended. Insufficient evidence exists to determine the effectiveness of IV iron in this patient group. CONCLUSION We advocate early consideration and investigation of anaemia, guided by symptom burden and patient preferences. Correction of reversible causes should be the mainstay of treatment, with a restrictive approach to blood transfusion. Research is required to evaluate the efficacy of IV iron in these patients.
Collapse
Affiliation(s)
- Karen Neoh
- St Gemma’s Hospice, Leeds, UK
- Karen Neoh, St Gemma’s Hospice, 329
Harrogate Road, Leeds, LS17 6QD, UK.
| | - Andrew Page
- Academic Unit of Palliative Care, Leeds
Institute of Health Sciences, Leeds, UK
| | - Nicolas Chin-Yee
- Department of Medicine, Division of
Palliative Care, St. Michael’s Hospital, Toronto, Canada
| | - Carolyn Doree
- NHS Blood and Transplant Systematic
Review Initiative, Oxford, UK
| | - Michael I Bennett
- Academic Unit of Palliative Care, Leeds
Institute of Health Sciences, Leeds, UK
| |
Collapse
|
140
|
Alnuwaysir RIS, Grote Beverborg N, van der Meer P. Fluctuating iron levels in heart failure: when and where to look at? Eur J Heart Fail 2022; 24:818-820. [PMID: 35415951 DOI: 10.1002/ejhf.2500] [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: 01/30/2022] [Revised: 04/04/2022] [Accepted: 04/10/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- R I S Alnuwaysir
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - N Grote Beverborg
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - P van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
141
|
Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure. J Am Coll Cardiol 2022; 79:e263-e421. [PMID: 35379503 DOI: 10.1016/j.jacc.2021.12.012] [Citation(s) in RCA: 1257] [Impact Index Per Article: 419.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM The "2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure" replaces the "2013 ACCF/AHA Guideline for the Management of Heart Failure" and the "2017 ACC/AHA/HFSA Focused Update of the 2013 ACCF/AHA Guideline for the Management of Heart Failure." The 2022 guideline is intended to provide patient-centric recommendations for clinicians to prevent, diagnose, and manage patients with heart failure. METHODS A comprehensive literature search was conducted from May 2020 to December 2020, encompassing studies, reviews, and other evidence conducted on human subjects that were published in English from MEDLINE (PubMed), EMBASE, the Cochrane Collaboration, the Agency for Healthcare Research and Quality, and other relevant databases. Additional relevant clinical trials and research studies, published through September 2021, were also considered. This guideline was harmonized with other American Heart Association/American College of Cardiology guidelines published through December 2021. STRUCTURE Heart failure remains a leading cause of morbidity and mortality globally. The 2022 heart failure guideline provides recommendations based on contemporary evidence for the treatment of these patients. The recommendations present an evidence-based approach to managing patients with heart failure, with the intent to improve quality of care and align with patients' interests. Many recommendations from the earlier heart failure guidelines have been updated with new evidence, and new recommendations have been created when supported by published data. Value statements are provided for certain treatments with high-quality published economic analyses.
Collapse
|
142
|
McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, Burri H, Butler J, Čelutkienė J, Chioncel O, Cleland JG, Coats AJ, Crespo-Leiro MG, Farmakis D, Gilard M, Heyman S, Hoes AW, Jaarsma T, Jankowska EA, Lainscak M, Lam CS, Lyon AR, McMurray JJ, Mebazaa A, Mindham R, Muneretto C, Francesco Piepoli M, Price S, Rosano GM, Ruschitzka F, Skibelund AK. Guía ESC 2021 sobre el diagnóstico y tratamiento de la insuficiencia cardiaca aguda y crónica. Rev Esp Cardiol 2022. [DOI: 10.1016/j.recesp.2021.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
143
|
Soto ME, Pérez-Torres I, Rubio-Ruiz ME, Manzano-Pech L, Guarner-Lans V. Interconnection between Cardiac Cachexia and Heart Failure—Protective Role of Cardiac Obesity. Cells 2022; 11:cells11061039. [PMID: 35326490 PMCID: PMC8946995 DOI: 10.3390/cells11061039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/25/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Cachexia may be caused by congestive heart failure, and it is then called cardiac cachexia, which leads to increased morbidity and mortality. Cardiac cachexia also worsens skeletal muscle degradation. Cardiac cachexia is the loss of edema-free muscle mass with or without affecting fat tissue. It is mainly caused by a loss of balance between protein synthesis and degradation, or it may result from intestinal malabsorption. The loss of balance in protein synthesis and degradation may be the consequence of altered endocrine mediators such as insulin, insulin-like growth factor 1, leptin, ghrelin, melanocortin, growth hormone and neuropeptide Y. In contrast to many other health problems, fat accumulation in the heart is protective in this condition. Fat in the heart can be divided into epicardial, myocardial and cardiac steatosis. In this review, we describe and discuss these topics, pointing out the interconnection between heart failure and cardiac cachexia and the protective role of cardiac obesity. We also set the basis for possible screening methods that may allow for a timely diagnosis of cardiac cachexia, since there is still no cure for this condition. Several therapeutic procedures are discussed including exercise, nutritional proposals, myostatin antibodies, ghrelin, anabolic steroids, anti-inflammatory substances, beta-adrenergic agonists, medroxyprogesterone acetate, megestrol acetate, cannabinoids, statins, thalidomide, proteasome inhibitors and pentoxifylline. However, to this date, there is no cure for cachexia.
Collapse
Affiliation(s)
- María Elena Soto
- Department of Immunology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico;
| | - Israel Pérez-Torres
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (I.P.-T.); (L.M.-P.)
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico;
| | - Linaloe Manzano-Pech
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (I.P.-T.); (L.M.-P.)
| | - Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico;
- Correspondence:
| |
Collapse
|
144
|
Martens P. The Effect of Iron Deficiency on Cardiac Function and Structure in Heart Failure with Reduced Ejection Fraction. Card Fail Rev 2022; 8:e06. [PMID: 35399547 PMCID: PMC8977990 DOI: 10.15420/cfr.2021.26] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/04/2021] [Indexed: 12/05/2022] Open
Abstract
Over the past decade, the detrimental impact of iron deficiency in heart failure with reduced ejection fraction has become abundantly clear, showing a negative impact on functional status, quality of life, cardiac function and structure, exercise capacity and an increased risk of hospitalisation due to heart failure. Mechanistic studies have shown the impact of iron deficiency in altering mitochondrial function and negatively affecting the already altered cardiac energetics in heart failure with reduced ejection fraction. Such failing energetics form the basis of the alterations to cellular myocyte shortening, culminating in reduced systolic function and cardiac performance. The IRON-CRT trials show that ferric carboxymaltose is capable of improving cardiac structure and cardiac performance. This article discusses the effect of iron deficiency on cardiac function and structure and how it can be alleviated.
Collapse
Affiliation(s)
- Pieter Martens
- Kauffman Center for Heart Failure Treatment and Recovery, Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, US
| |
Collapse
|
145
|
Pereira GAR, Beck-da-Silva L. Deficiência de Ferro na Insuficiência Cardíaca com Fração de Ejeção Reduzida: Fisiopatologia, Diagnóstico e Tratamento. Arq Bras Cardiol 2022; 118:646-654. [PMID: 35319614 PMCID: PMC8959039 DOI: 10.36660/abc.20201257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/12/2021] [Indexed: 11/18/2022] Open
Abstract
A deficiência de ferro (DF) ou ferropenia é uma importante comorbidade na insuficiência cardíaca com fração de ejeção reduzida (ICFER) estável, e muito prevalente tanto nos anêmicos como não anêmicos. A ferropenia na ICFER deve ser pesquisada por meio da coleta de saturação de transferrina e ferritina. Há dois tipos de ferropenia na IC: absoluta, em que as reservas de ferro estão depletadas; e funcional, onde o suprimento de ferro é inadequado apesar das reservas normais. A ferropenia está associada com pior classe funcional e maior risco de morte em pacientes com ICFER, e evidências científicas apontam melhora de sintomas e de qualidade de vida desses pacientes com tratamento com ferro parenteral na forma de carboximaltose férrica. O ferro exerce funções imprescindíveis como o transporte (hemoglobina) e armazenamento (mioglobina) de oxigênio, além de ser fundamental para o funcionamento das mitocôndrias, constituídas de proteínas à base de ferro, e local de geração de energia na cadeia respiratória pelo metabolismo oxidativo. A geração insuficiente e utilização anormal de ferro nas células musculares esquelética e cardíaca contribuem para a fisiopatologia da IC. A presente revisão tem o objetivo de aprofundar o conhecimento a respeito da fisiopatologia da ferropenia na ICFER, abordar as ferramentas disponíveis para o diagnóstico e discutir sobre a evidência científica existente de reposição de ferro.
Collapse
|
146
|
Gevaert AB, Mueller S, Winzer EB, Duvinage A, Van de Heyning CM, Pieske-Kraigher E, Beckers PJ, Edelmann F, Wisløff U, Pieske B, Adams V, Halle M, Van Craenenbroeck EM, for the OptimEx-Clin Study Group. Iron Deficiency Impacts Diastolic Function, Aerobic Exercise Capacity, and Patient Phenotyping in Heart Failure With Preserved Ejection Fraction: A Subanalysis of the OptimEx-Clin Study. Front Physiol 2022; 12:757268. [PMID: 35222057 PMCID: PMC8866976 DOI: 10.3389/fphys.2021.757268] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/27/2021] [Indexed: 01/08/2023] Open
Abstract
Aims Iron deficiency (ID) is linked to reduced aerobic exercise capacity and poor prognosis in patients with heart failure (HF) with reduced ejection fraction (HFrEF); however, data for HF with preserved ejection fraction (HFpEF) is scarce. We assessed the relationship between iron status and diastolic dysfunction as well as aerobic exercise capacity in HFpEF, and the contribution of iron status to patient phenotyping. Methods and Results Among 180 patients with HFpEF (66% women; median age, 71 years) recruited for the Optimizing Exercise Training in Prevention and Treatment of Diastolic HF (OptimEx-Clin) trial, baseline iron status, including iron, ferritin, and transferrin saturation, was analyzed (n = 169) in addition to exercise capacity (peak oxygen uptake [peak V̇O2]) and diastolic function (E/e′). ID was present in 60% of patients and was more common in women. In multivariable linear regression models, we found that diastolic function and peak V̇O2 were independently related to iron parameters; however, these relationships were present only in patients with HFpEF and ID [E/e′ and iron: β−0.19 (95% confidence interval −0.32, −0.07), p = 0.003; E/e′ and transferrin saturation: β−0.16 (−0.28, −0.04), p = 0.011; peak V̇O2 and iron: β 3.76 (1.08, 6.44), p = 0.007; peak V̇O2 and transferrin saturation: β 3.58 (0.99, 6.16), p = 0.007]. Applying machine learning, patients were classified into three phenogroups. One phenogroup was predominantly characterized by the female sex and few HFpEF risk factors but a high prevalence of ID (86%, p < 0.001 vs. other phenogroups). When excluding ID from the phenotyping analysis, results were negatively influenced. Conclusion Iron parameters are independently associated with impaired diastolic function and low aerobic capacity in patients with HFpEF and ID. Patient phenotyping in HFpEF is influenced by including ID. Clinical Trial Registration www.ClinicalTrials.gov, identifier NCT02078947.
Collapse
Affiliation(s)
- Andreas B. Gevaert
- Research Group Cardiovascular Diseases, GENCOR (Genetics, Pharmacology & Physiopathology of Heart, Blood, Vessels and Skeleton) Department, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
- *Correspondence: Andreas B. Gevaert,
| | - Stephan Mueller
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Ephraim B. Winzer
- Department of Internal Medicine and Cardiology, Heart Center Dresden – University Hospital, Technische Universität Dresden, Dresden, Germany
| | - André Duvinage
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Caroline M. Van de Heyning
- Research Group Cardiovascular Diseases, GENCOR (Genetics, Pharmacology & Physiopathology of Heart, Blood, Vessels and Skeleton) Department, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Elisabeth Pieske-Kraigher
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Paul J. Beckers
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Ulrik Wisløff
- Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Volker Adams
- Department of Internal Medicine and Cardiology, Heart Center Dresden – University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR (Genetics, Pharmacology & Physiopathology of Heart, Blood, Vessels and Skeleton) Department, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | | |
Collapse
|
147
|
Kozłowska B, Sochanowicz B, Kraj L, Palusińska M, Kołsut P, Szymański Ł, Lewicki S, Śmigielski W, Kruszewski M, Leszek P. Expression of Iron Metabolism Proteins in Patients with Chronic Heart Failure. J Clin Med 2022; 11:jcm11030837. [PMID: 35160288 PMCID: PMC8837054 DOI: 10.3390/jcm11030837] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 11/25/2022] Open
Abstract
In heart failure, iron deficiency is a common comorbid disease that negatively influences exercise tolerance, number of hospitalizations and mortality rate, and this is why iron iv supplementation is recommended. Little is known about the changes in iron-related proteins in the human HF myocardium. The purpose of this study was to assess iron-related proteins in non-failing (NFH) vs. failing (FH) human myocardium. The study group consisted of 58 explanted FHs; control consisted of 31 NFHs unsuitable for transplantation. Myocardial proteins expressions: divalent metal transporter (DMT-1); L-type calcium channel (L-CH); transferrin receptors (TfR-1/TfR-2); ferritins: heavy (FT-H) or light (FT-L) chain, mitochondrial (FT-MT); ferroportin (FPN), regulatory factors and oxidative stress marker: 4-hydroxynonenal (4-HNE). In FH, the expression in almost all proteins responsible for iron transport: DMT-1, TfR-1, L-CH, except TfR-2, and storage: FT-H/-L/-MT were reduced, with no changes in FPN. Moreover, 4-HNE expression (pg/mg; NFH 10.6 ± 8.4 vs. FH 55.7 ± 33.7; p < 0.0001) in FH was increased. HNE-4 significantly correlated with DMT-1 (r = −0.377, p = 0.036), L-CH (r = −0.571, p = 0.001), FT-H (r = −0.379, p = 0.036), also FPN (r = 0.422, p = 0.018). Reducing iron-gathering proteins and elevated oxidative stress in failing hearts is very unfavorable for myocardiocytes. It should be taken into consideration before treatment with drugs or supplements that elevate free oxygen radicals in the heart.
Collapse
Affiliation(s)
- Bogna Kozłowska
- Department of Heart Failure and Transplantology, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
| | - Barbara Sochanowicz
- Centre of Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warszawa, Poland; (B.S.); (M.K.)
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, 01-163 Warsaw, Poland;
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
| | - Małgorzata Palusińska
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
| | - Piotr Kołsut
- Department of Cardiac Surgery and Transplantology, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
| | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
| | - Sławomir Lewicki
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
- Faculty of Medical Sciences and Health Sciences, Kazimierz Pulaski University of Technology and Humanities, 26-600 Radom, Poland
| | - Witold Śmigielski
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
| | - Marcin Kruszewski
- Centre of Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warszawa, Poland; (B.S.); (M.K.)
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| | - Przemysław Leszek
- Department of Heart Failure and Transplantology, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-3434-483
| |
Collapse
|
148
|
Campodonico J, Junod D, Carulli E, Lo Russo G, Gaudenzi Asinelli M, Doni F, Bonomi A, Agostoni P. Role of impaired iron transport on exercise performance in heart failure patients. Eur J Prev Cardiol 2022; 29:1104-1111. [PMID: 35134891 DOI: 10.1093/eurjpc/zwab216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/30/2021] [Indexed: 12/28/2022]
Abstract
AIMS Impaired iron transport (IIT) occurs frequently in heart failure (HF) patients, even in the absence of anaemia and it is associated with a poor quality of life and prognosis. The impact of IIT on exercise capacity, as assessed by the cardiopulmonary exercise test (CPET), in HF is at present unknown. The aim of this article is to evaluate in HF patients the impact on exercise performance of IIT, defined as transferrin saturation (TSAT) <20%. METHODS AND RESULTS We collected data of 676 patients hospitalized for HF. All underwent laboratory analysis, cardiac ultrasound, and CPET. Patients were grouped by the presence/absence of IIT and anaemia (haemoglobin <13 and <12 g/dL in male and female, respectively): Group 1 (G1) no anaemia, no IIT; Group 2 (G2) anaemia, no IIT; Group 3 (G3) no anaemia, IIT; Group 4 (G4) anaemia and IIT. Peak oxygen uptake (peakVO2) reduced from G1 to G3 and from G2 to G4 (G1: 1266 ± 497 mL/min, G2: 1011 ± 385 mL/min, G3: 1041 ± 395 mL/min, G4: 833 ± 241 mL/min), whereas the ventilation to carbon dioxide relationship slope (VE/VCO2 slope) increased (G1: 31.8 ± 7.5, G2: 34.5 ± 7.4, G3: 36.1 ± 10.2, G4: 37.5 ± 8.4). At multivariate regression analysis, peakVO2 independent predictors were anaemia, brain natriuretic peptide (BNP), and left ventricular ejection fraction, whereas VE/VCO2 slope independent predictors were IIT and BNP. CONCLUSION In HF IIT is associated with exercise performance impairment independently from anaemia, and it is a predictor of elevated VE/VCO2 slope, a pivotal index of HF prognosis.
Collapse
Affiliation(s)
- Jeness Campodonico
- Centro Cardiologico Monzino, IRCCS, Via Parea 4, 20138 Milano, Italy.,Translational Medicine PhD Course, University of Milan, Milan, Italy
| | - Daniele Junod
- Cardiovascular Section, Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Ermes Carulli
- Cardiovascular Section, Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Gerardo Lo Russo
- Cardiovascular Section, Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | | | - Francesco Doni
- Cardiovascular Section, Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Alice Bonomi
- Centro Cardiologico Monzino, IRCCS, Via Parea 4, 20138 Milano, Italy
| | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCS, Via Parea 4, 20138 Milano, Italy.,Cardiovascular Section, Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
149
|
McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, Burri H, Butler J, Čelutkienė J, Chioncel O, Cleland JGF, Coats AJS, Crespo-Leiro MG, Farmakis D, Gilard M, Heymans S, Hoes AW, Jaarsma T, Jankowska EA, Lainscak M, Lam CSP, Lyon AR, McMurray JJV, Mebazaa A, Mindham R, Muneretto C, Francesco Piepoli M, Price S, Rosano GMC, Ruschitzka F, Kathrine Skibelund A. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: Developed by the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). With the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail 2022; 24:4-131. [PMID: 35083827 DOI: 10.1002/ejhf.2333] [Citation(s) in RCA: 1221] [Impact Index Per Article: 407.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022] Open
Abstract
Document Reviewers: Rudolf A. de Boer (CPG Review Coordinator) (Netherlands), P. Christian Schulze (CPG Review Coordinator) (Germany), Magdy Abdelhamid (Egypt), Victor Aboyans (France), Stamatis Adamopoulos (Greece), Stefan D. Anker (Germany), Elena Arbelo (Spain), Riccardo Asteggiano (Italy), Johann Bauersachs (Germany), Antoni Bayes-Genis (Spain), Michael A. Borger (Germany), Werner Budts (Belgium), Maja Cikes (Croatia), Kevin Damman (Netherlands), Victoria Delgado (Netherlands), Paul Dendale (Belgium), Polychronis Dilaveris (Greece), Heinz Drexel (Austria), Justin Ezekowitz (Canada), Volkmar Falk (Germany), Laurent Fauchier (France), Gerasimos Filippatos (Greece), Alan Fraser (United Kingdom), Norbert Frey (Germany), Chris P. Gale (United Kingdom), Finn Gustafsson (Denmark), Julie Harris (United Kingdom), Bernard Iung (France), Stefan Janssens (Belgium), Mariell Jessup (United States of America), Aleksandra Konradi (Russia), Dipak Kotecha (United Kingdom), Ekaterini Lambrinou (Cyprus), Patrizio Lancellotti (Belgium), Ulf Landmesser (Germany), Christophe Leclercq (France), Basil S. Lewis (Israel), Francisco Leyva (United Kingdom), AleVs Linhart (Czech Republic), Maja-Lisa Løchen (Norway), Lars H. Lund (Sweden), Donna Mancini (United States of America), Josep Masip (Spain), Davor Milicic (Croatia), Christian Mueller (Switzerland), Holger Nef (Germany), Jens-Cosedis Nielsen (Denmark), Lis Neubeck (United Kingdom), Michel Noutsias (Germany), Steffen E. Petersen (United Kingdom), Anna Sonia Petronio (Italy), Piotr Ponikowski (Poland), Eva Prescott (Denmark), Amina Rakisheva (Kazakhstan), Dimitrios J. Richter (Greece), Evgeny Schlyakhto (Russia), Petar Seferovic (Serbia), Michele Senni (Italy), Marta Sitges (Spain), Miguel Sousa-Uva (Portugal), Carlo G. Tocchetti (Italy), Rhian M. Touyz (United Kingdom), Carsten Tschoepe (Germany), Johannes Waltenberger (Germany/Switzerland) All experts involved in the development of these guidelines have submitted declarations of interest. These have been compiled in a report and published in a supplementary document simultaneously to the guidelines. The report is also available on the ESC website www.escardio.org/guidelines For the Supplementary Data which include background information and detailed discussion of the data that have provided the basis for the guidelines see European Heart Journal online.
Collapse
|
150
|
Hematopoiesis, Inflammation and Aging-The Biological Background and Clinical Impact of Anemia and Increased C-Reactive Protein Levels on Elderly Individuals. J Clin Med 2022; 11:jcm11030706. [PMID: 35160156 PMCID: PMC8836692 DOI: 10.3390/jcm11030706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Anemia and systemic signs of inflammation are common in elderly individuals and are associated with decreased survival. The common biological context for these two states is then the hallmarks of aging, i.e., genomic instability, telomere shortening, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intercellular communication. Such aging-associated alterations of hematopoietic stem cells are probably caused by complex mechanisms and depend on both the aging of hematopoietic (stem) cells and on the supporting stromal cells. The function of inflammatory or immunocompetent cells is also altered by aging. The intracellular signaling initiated by soluble proinflammatory mediators (e.g., IL1, IL6 and TNFα) is altered during aging and contributes to the development of both the inhibition of erythropoiesis with anemia as well as to the development of the acute-phase reaction as a systemic sign of inflammation with increased CRP levels. Both anemia and increased CRP levels are associated with decreased overall survival and increased cardiovascular mortality. The handling of elderly patients with inflammation and/or anemia should in our opinion be individualized; all of them should have a limited evaluation with regard to the cause of the abnormalities, but the extent of additional and especially invasive diagnostic evaluation should be based on an overall clinical evaluation and the possible therapeutic consequences.
Collapse
|