You are on page 1of 24

nature reviews cardiology https://doi.org/10.

1038/s41569-024-00988-1

Review article Check for updates

Iron deficiency and


supplementation in heart failure
Samira Lakhal-Littleton 1
& John G. F. Cleland 2

Abstract Sections

Non-anaemic iron deficiency (NAID) is a strategic target in cardiovascular Introduction

medicine because of its association with a range of adverse effects Iron and its markers
in various conditions. Endeavours to tackle NAID in heart failure Relationship between iron
have yielded mixed results, exposing knowledge gaps in how best to status and HF
define ‘iron deficiency’ and the handling of iron therapies by the body. Iron supplementation in HF
To address these gaps, we harness the latest understanding of the
Opportunities in the
mechanisms of iron homeostasis outside the erythron and integrate management of ID in HF
clinical and preclinical lines of evidence. The emerging picture is that Conclusions
current definitions of iron deficiency do not assimilate the multiple
influences at play in patients with heart failure and, consequently, fail
to identify those with a truly unmet need for iron. Additionally, current
iron supplementation therapies benefit only certain patients with
heart failure, reflecting differences in the nature of the unmet need
for iron and the modifying effects of anaemia and inflammation on
the handling of iron therapies by the body. Building on these insights,
we identify untapped opportunities in the management of NAID,
including the refinement of current approaches and the development
of novel strategies. Lessons learned from NAID in cardiovascular
disease could ultimately translate into benefits for patients with other
chronic conditions such as chronic kidney disease, chronic obstructive
pulmonary disease and cancer.

Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK. 2British Heart Foundation
1

Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK.
e-mail: samira.lakhal-littleton@dpag.ox.ac.uk

Nature Reviews Cardiology


Review article

Key points Iron and its markers


Iron regulation in the body
Iron is an essential trace element found in all living organisms. The aver-
•• Non-anaemic iron deficiency (NAID) is a strategic target in age amount of iron in the human body starts at ~0.2 g at birth, increasing
cardiovascular medicine because of its high prevalence, adverse throughout childhood to ~3.0 g, then plateauing in adulthood at ~3.5 g
effects on a range of outcomes and its role as a precursor to anaemia. in women and ~4.0 g in men15–18. Iron fluxes between oxidative states,
complexing ligands and functional pools are shown in Fig. 2.
•• In patients with heart failure (HF), variation in iron markers reflects Iron can exist in various oxidation states in the body, most fre-
the demographic factors that influence them in the general population quently as ferrous (Fe2+) or ferric (Fe3+) iron19,20. In the gut lumen, Fe3+ is
as well as the modifying effects of common comorbidities and reduced to Fe2+ by plasma membrane ascorbate-dependent reductase
certain medications. CYBRD1 (also known as duodenal cytochrome b) to allow uptake by the
divalent metal transporter 1 (DMT1)21. The iron exporter solute carrier
•• Mechanisms underlying the adverse effects of NAID in HF could family 40 member 1 (also known as ferroportin) exports Fe2+ from duo-
include the unmet needs for iron by the myocardium, skeletal muscle denal enterocytes, hepatocytes or splenic macrophages, which are the
or pulmonary vasculature and the role of iron dysregulation in cells involved in iron absorption, storage and recycling, respectively.
comorbidities. Fe2+ is then oxidized back to Fe3+ by hephaestin or ceruloplasmin, before
being loaded onto the plasma iron chaperone transferrin22.
•• The benefits of oral iron therapies should be re-examined in light Iron forms complexes with various ligands. In the circulation, iron
of the latest developments in dosing regimens and slow-release can be tightly bound to its chaperone transferrin in the Fe3+ form22,23
formulations. and can also be found as non-transferrin-bound iron (NTBI), typically
as citrate-bound Fe3+ or albumin-bound Fe2+ or Fe3+ (ref. 19). Inside
•• Although intravenous iron therapy benefits some patients with HF, cells, ~40–50% of iron is bound to the cellular storage protein ferritin
the fate of iron in the body and the long-term safety of repeated dosing as Fe3+, and ~40–50% is contained within prosthetic groups such as
remain unknown. haem (as Fe2+) or iron–sulfur groups (as Fe2+or Fe3+) mostly in mito-
chondrial enzymes4,5,15,24,25. In cardiac and skeletal muscle cells, iron
•• Identifying markers of the unmet need for iron by tissues and is also contained in the haem group of the oxygen storage protein
targeting the iron homeostatic pathways in the body hold the potential myoglobin26. Only a small proportion of cellular iron, typically <5%,
to transform the management of NAID. is labile (termed the labile iron pool (LIP)), where iron is either ligand
free, bound to reduced glutathione or coordinated by iron chaperones
such as poly(rC)-binding proteins19,20,22.
Introduction Iron also participates in various ‘functional’ pools. The largest,
The haemoglobin-centric view has long dominated our understand- accounting for around two-thirds of total iron in the body, is the eryth-
ing of the adverse health effects of iron deficiency (ID). In patients roid pool in which the primary function of iron is to bind oxygen to the
with cardiovascular disease (CVD), the haemoglobin level is often haem prosthetic group of haemoglobin for oxygen transport around
measured as a proxy for iron status1–3. However, haemoglobin is only the body27. Pro-erythroblasts and erythroblasts take up ~80% of circu-
one of many life-sustaining proteins whose function depends on iron4,5 lating iron daily via transferrin receptor protein 1 (TFR1), then shed any
(Fig. 1). Impairment of haemoglobin synthesis is a late consequence remaining receptors into the circulation in soluble form (sTFR)28. The
of ID, with anaemia manifesting in only one-third of iron-deficient erythroid pool accounts for most of the daily iron demand and there-
individuals6. Non-anaemic ID (NAID) has long been overlooked but fore exerts dominant control over systemic iron homeostasis29,30. Iron is
is now recognized as a strategic clinical target in patients with heart recycled from the erythroid pool by reticuloendothelial macrophages
failure (HF) because of its high prevalence, its adverse effects on in the spleen following the breakdown of senescent red blood cells27.
multiple outcomes and its role as a precursor to the comorbidity of Hepatocytes contain the second largest functional pool, in which
anaemia3,7–14 (Table 1). iron is stored in an inert form in ferritin shells (each of which can hold
Recognition of NAID as a strategic clinical target has coincided up to 4,500 atoms) but can be readily mobilized to meet the demands
with a paradigm shift in approaches to iron replacement, from incre- of the erythron, growth or pregnancy24. The iron content of the liver
mental oral therapies to bolus intravenous therapies. Cardiovascular at birth is in the order of ~30 mg, rising in adulthood to ~1,000 mg
medicine has been at the leading edge of these developments, both in in men and ~300 mg in women31,32. When the rate of iron entry into
adopting intravenous iron therapies and in recognizing the potential the plasma exceeds the rate of its clearance by the liver, NTBI levels
gains of treating NAID in patients with CVD. However, endeavours to increase and iron is deposited in secondary storage organs, such as the
unlock that potential have been met with challenges, exposing the heart and pancreas, owing to their high expression of NTBI transport-
substantial limitations of current approaches to the diagnosis and ers, including DMT1, zinc transporters 8 and 14, and L-type calcium
management of ID. channels20,33,34.
In this Review, we harness the latest understanding of iron home- The third largest functional pool is the intracellular pool in
ostasis to evaluate existing and emerging markers of iron status, non-erythroid tissues, where the primary function of iron is to activate
interrogate the reciprocal relationship between iron status and HF, oxygen for a wide range of cellular chemistries. For example, iron in
and mechanistically examine the benefits of oral and intravenous haem and iron–sulfur groups catalyses the activation of oxygen for oxi-
iron replacement therapies. We conclude by highlighting a range dative phosphorylation, and unbound iron in the LIP activates oxygen
of untapped opportunities to improve the management of NAID in via cytoplasmic dioxygenases4,5,22. The cellular LIP is controlled at the
patients with HF. point of uptake (TFR1) and storage (cellular ferritin) and, in some cells

Nature Reviews Cardiology


Review article

DNA replication and repair Myelin biosynthesis


XPD Stearoyl-CoA ∆9-desaturase MUFAs
Polα/δ/ε Palmitoyl-CoA

Polα/δ/ε Trimethyl lysine TMLH GBBH Carnitine


NTHL1
Lanosterol CYP51A1 Cholesterol
Cellular respiration
Neurotransmitter release
CI CII CIII CIV
Phenylalanine PAH Tyrosine TH Dopamine
CytoC
Tryptophan TPH Serotonin
NADH FAD
NAD+ FADH2 Oxygen storage
SD

Krebs O2
H

cycle
Deoxymyoglobin Oxymyoglobin
CO
A

1 FH O2

Oxygen sensing Vitamin D metabolism


PHD1/2/3 Degradation Calcitoic
HIF1/2α Vitamin D CYP2R1 25(OH)D CYP27B1 1,25(OH)2D CYP24A1 acid
FIH Inactivation

Oxygen transport
Collagen synthesis
4 × O2
Р4Н
Procollagen Mature collagen Deoxyhaemoglobin Oxyhaemoglobin
LH 4 × O2

Fe Haem Fe Fe–S cluster

Fig. 1 | Vital iron-dependent proteins and their signalling pathways. cytochrome c; FH, fumarate hydratase; FIH, hypoxia-inducible factor 1α inhibitor
Proteins requiring free iron are shown in yellow. Proteins requiring iron within (also known as factor inhibiting HIF1α); GBBH, γ-butyrobetaine dioxygenase; HIF,
a haem functional group are shown in red. Proteins requiring iron within an hypoxia-inducible factor; LH, lysyl hydroxylase; MUFA, monounsaturated fatty
Fe–S functional group are shown in blue. Only iron-dependent steps are shown. acid; NTHL1, endonuclease III-like protein 1 (also known as DNA glycosylase);
1,25(OH)2D, 1,25-dihydroxyvitamin D; 25(OH)D, 25-hydroxyvitamin D; ACO1, P4H, prolyl 4-hydroxylase; PAH, phenylalanine-4-hydroxylase; PHD1, prolyl
aconitate hydratase 1; CI, complex I; CII, complex II; CIII, complex III; CIV, hydroxylase 1; Polα, polymerase subunit-α; SDH, succinate dehydrogenase;
complex IV; CYP2R1, vitamin D 25-hydroxylase; CYP24A1, 1,25-dihydroxyvitamin TH, tyrosine 3-monooxygenase (also known as tyrosine hydroxylase); TPH,
D3 24-hydroxylase, mitochondrial; CYP27B1, 25-hydroxyvitamin D-1α tryptophan hydroxylase; TMLH, trimethyllysine dioxygenase; XPD, helicase
hydroxylase, mitochondrial; CYP51A1, lanosterol 14α demethylase; CytoC, subunit of transcription factor IIH.

(such as cardiomyocytes and vascular smooth muscle cells), addi- No active processes exist for iron excretion from the body because
tionally via ferroportin-mediated export23,35–38. Despite constituting all the iron that is filtered through the kidneys is reabsorbed48,49. A small
<5% of intracellular iron, the LIP is important as the substrate that is amount of iron (~1–2 mg daily) is lost through the sloughing of skin
sensed by iron regulatory proteins (IRPs) that ultimately orchestrate and mucosal cells46,47. In menstruating women, up to 50 mg of iron is
cellular uptake, usage and storage, as an obligatory intermediate in the lost per cycle50,51 and, in pregnancy, >500 mg of maternal iron is irre-
exchange of iron between ligands, and as the cause of tissue damage versibly ‘lost’ to fetal development52,53. The typical 100 mg oral iron
when in excess19,32,35,39–41. dose results in about 10 mg being absorbed into the circulation54,55.
Another functional pool is the circulating iron pool, in which ~3 mg Transfusion delivers ~200 mg of iron per unit of blood into the eryth-
of iron is bound to transferrin, with NTBI becoming detectable when roid pool56. Intravenous iron formulations release some iron directly
transferrin saturation (Tsat) exceeds 70%22,32,42. Although ferritin mole­ into the circulating pool as NTBI and some indirectly following
cules are abundant in the circulation, they are iron-poor and derive macrophage-dependent release of iron from the carbohydrate shell,
primarily from macrophages rather than hepatocytes43–45. Iron enters with the proportion of iron released as NTBI ranging between 10% and
the circulating pool following ferroportin-mediated export from gut 40% depending on the formulation57,58.
enterocytes, reticuloendothelial macrophages and hepatocytes46,47.
Ferroportin at these sites is subject to post-translational control by Circulating iron markers
the iron homeostatic hormone hepcidin, which causes its internaliza- Circulating iron markers reflect the iron homeostatic pathways that
tion and degradation46,47. The circulating iron pool is important as govern them and, in disease, they also indicate the influence of inflamma-
the substrate of iron sensing that sets hepcidin levels, an obligatory tion and hypoxia. The profound effects of inflammation on iron markers
intermediate in the influx of dietary and parenteral iron and in the originated in the evolutionary ‘arms race’ between host and pathogens
exchange of iron between functional pools as well as being the primary for access to iron, meaning that the signals that accompany infection
site of sampling in the clinical assessment of iron status. (and sterile inflammation) also influence iron handling in the body59,60.

Nature Reviews Cardiology


Review article

Table 1 | Prevalence and outcomes of NAID in studies of patients with HF

Patients Number Definition of NAID Prevalence of Clinical outcomes associated with NAID Ref.
NAID (%)

HFpEF (LVEF ≥50%); HFmrEF 15,197 Ferritin <100 μg/l or ferritin HFpEF 61.0, HFmrEF Increased incidence of recurrent hospitalization for 3
(LVEF 40–49%); HFrEF (LVEF <40%) 100–299 μg/l with Tsat <20% 51.0, HFrEF 54.0 HF and cardiovascular death (HR 1.63)a
CHF with LVEF ≤45% 1,198 Ferritin <100 µg/l or ferritin 42.5 Reduced maximum exercise capacityb 7
100–299 µg/l with Tsat <20%
HFpEF (LVEF >45%) or HFrEF 1,506 Ferritin <100 µg/l or ferritin 50.0 All-cause mortality (HR 1.42)c 8
(LVEF ≤45%) 100–299 µg/l with Tsat <20%
AHF (discharged from hospital) 626 Ferritin <100 µg/l 48.2 Increased risk of 30-day hospital readmission 9
(HR 1.72)d
Ferritin 100–299 µg/l with 25.7 No effect on risk of 30-day hospital readmission
Tsat <20%
HFpEF (LVEF >50%); HFmrEF 1,197 Ferritin <100 µg/l or ferritin HFpEF 64.0, HFmrEF Reduced VO2e; freedom from all-cause death and 10
(LVEF 40–50%); HFrEF (LVEF <40%) 100–299 µg/l with Tsat <20% 61.0, HFrEF 50.0 heart failure hospitalizatione; progression from
NAID to IDA
Stable systolic CHF (LVEF 26 ± 7%) 546 Ferritin <100 µg/l or ferritin 37.0 Increased risk of death or heart transplantation 11
100–299 µg/l with Tsat <20% (HR 1.58)f
DHF 1,701 Ferritin <100 μg/l or ferritin 73.3 Increase in composite of 30-day death or hospital 12
100–299 μg/l with Tsat <20% readmission for DHF (HR 1.75; lower quartile Tsat
versus upper quartile Tsat)g
AHF, acute heart failure; CHF, chronic heart failure; DHF, decompensated heart failure; HF, heart failure; HFmrEF, heart failure with mid-range ejection fraction; HFpEF, heart failure with
preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; hsCRP, C-reactive protein level measured by high-sensitivity assay; IDA, iron deficiency anaemia; LVEF, left
ventricular ejection fraction; NAID, non-anaemic iron deficiency; Tsat, transferrin saturation; VO2, peak oxygen consumption. aAdjusted for centre, age, sex, BMI, CHF aetiology, NYHA class,
LVEF, plasma N-terminal pro-B-type natriuretic peptide (NT-proBNP) level, serum sodium level, serum hsCRP level, renal function assessed using estimated glomerular filtration rate, presence
of diabetes mellitus and presence of anaemia. bAdjusted for age, sex, BMI, diabetes status, NYHA functional class, LVEF, renal function, levels of hsCRP and NT-proBNP, treatment with
angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, statins or loop diuretics, and presence of anaemia. cAdjusted for prior admission to hospital for AHF, presence of
hypertension or dyslipidaemia, length of hospital stay, interaction between LVEF ≤50% and diastolic blood pressure, and NT-proBNP and troponin T (high-sensitivity assay) levels. dAdjusted for
age, sex, anaemia, LVEF, and levels of serum creatinine, CRP and natriuretic peptides. eAdjusted for age, sex, implantable cardioverter–defibrillator use, cardiac resynchronization therapy use,
ischaemic aetiology of HF, use of angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, β-blockers or mineralocorticoid receptor antagonists, NYHA functional class, loop
diuretic use, and baseline LVEF. fAdjusted for age, sex, last NYHA functional class III/IV under stable conditions, prior admission for DHF, presence of dyslipidaemia, Charlson comorbidity index,
systolic blood pressure, heart rate, atrial fibrillation, LVEF, left atrial diameter, haemoglobin, estimated glomerular filtration rate and NT-proBNP levels. gAdjusted for sociodemographic and
clinical factors, comorbidities, treatment variables, and anaemia.

The influence of hypoxia (anaemic hypoxia and hypoxaemia of severe tissues75. Transferrin serum levels depend on the rates of biosynthesis in
chronic obstructive pulmonary disease (COPD)) is underpinned by the liver, consumption for erythropoiesis and clearance by the kidneys42.
the extensive crosstalk between iron-sensing and oxygen-sensing Transferrin biosynthesis in the liver is increased by ID anaemia, hypoxia,
pathways4,5,61. Because inflammation and hypoxia are common denomi- or when liver iron stores are depleted (such as during pregnancy) and
nators in CVD, correct interpretation of circulating iron markers requires is decreased when liver iron stores are excessive or as a result of liver
a firm understanding of these influences (Fig. 3 and Table 2). injury42,76–80. Serum transferrin levels can also be reduced due to urinary
loss in patients with renal disease81,82. In acute settings, TNF suppresses
Ferritin. Ferritin is both an iron-storage protein and a positive acute- transferrin biosynthesis, and serum transferrin levels can drop mark-
phase protein. In comparison with intracellular ferritin, serum ferritin edly (for example, by ~30% after myocardial infarction)83–87. Raised
is iron-poor, has a high ratio of light-to-heavy-chain subunits and is transferrin levels can therefore denote the depletion of liver iron stores;
frequently glycosylated, with evidence that the level of glycosylation however, this rise would be masked by the opposing effects of inflamma-
is increased by inflammation45,62,63. Serum ferritin is primarily derived tion, liver injury, nephrosis and excessive erythropoiesis (for example,
from macrophages rather than hepatocytes, although hepatocyte ineffective erythropoiesis and erythroid anaemia).
ferritin can leak into the circulation as a result of liver injury43,44,64,65.
In macrophages, tumour necrosis factor (TNF) regulates the expression Serum iron. The serum iron measured in standard clinical tests consti-
of the ferritin heavy chain, whereas IRPs regulate the expression of both tutes the sum of transferrin-bound iron and NTBI. Serum iron levels are
heavy and light chains23,66–68. High serum ferritin levels are associated determined by the rates of export from supply tissues and uptake by
with high mortality in a broad range of underlying pathologies, includ- demand tissues19,47,76,80. The levels are also subject to diurnal variation,
ing myocardial infarction and HF69–74. Therefore, serum ferritin levels changing by as much as 30% during the course of the day88. A reduced
reflect the interaction between inflammation and iron in macrophages serum iron level can denote low iron stores in the body, increased
as well as the extent of liver injury in patients with liver disease. demand during growth or for erythropoiesis, or increased sequestra-
tion due to infection or inflammation (due to hepcidin elevation).
Transferrin. Transferrin is both a serum iron chaperone and a negative By contrast, elevated serum iron levels can denote excess iron stor-
acute-phase protein22,42. The importance of transferrin is demonstrated age, an acute response to hypoxia, reduced iron consumption by the
by the genetic disorder atransferrinaemia, which is characterized by bone marrow (such as with cytotoxic therapies) or failure to produce
both microcytic hypochromic anaemia and excess iron deposition in sufficient transferrin due to liver injury89–93.

Nature Reviews Cardiology


Review article

Transferrin saturation. Tsat represents the proportion of iron-binding somatic mutations have HIF2 accumulation, excess erythropoietin
sites in transferrin molecules that are occupied by iron and is often production, stress erythropoiesis, polycythaemia and suppressed
calculated using serum iron and total iron-binding capacity, which hepcidin levels121,122. TNF decreases renal erythropoietin production
reflects serum transferrin concentration94. Therefore, the interpreta- and suppresses differentiation of erythroid progenitors in the bone
tion of changes in Tsat should consider both the factors that influence marrow, whereas IFNγ reduces the lifespan of red blood cells, speed-
serum iron levels and those that influence serum transferrin levels. ing up their turnover123,124. Under these conditions, the body resorts to
stress erythropoiesis in the spleen driven by growth/differentiation
Soluble TFR. sTFR derives primarily from the shedding of TFR1 from factor 15 (ref. 125). Therefore, ACD is not solely the consequence of
erythroblasts95–97. IRPs upregulate TFR1 expression in iron-hungry hepcidin-driven iron sequestration but also the direct influence
erythroblasts and, therefore, sTFR levels reflect not only the size of of inflammation on erythropoietin. Furthermore, sampling the bone
the erythroblast pool but also its demand for iron. Indeed, the sTFR marrow in inflammatory settings might not produce a complete picture
level is increased in β-thalassaemia and in response to erythropoietin of erythroid iron demand, because of stress erythropoiesis.
dose, and is reduced by bone marrow suppression (such as with chemo-
therapy)98–100. sTFR levels have not been reported to be influenced by Definition and diagnosis of ID
inflammation; consequently, sTFR and the sTFR/log ferritin index have The management of ID faces two challenges. First, guidelines for the
been proposed as superior diagnostic markers for ID anaemia101–103. management of ID126–131 are still intertwined with those for anaemia,
meaning that NAID is often not screened for and iron markers are inves-
Hepcidin. Serum hepcidin is the master hormone of systemic iron tigated only when anaemia is suspected based on full blood count or
homeostasis and derives primarily from hepatocytes46,91. Some cells haemoglobin measurements. For instance, in patients with HF, iron
(including cardiomyocytes) also express hepcidin although, in this markers are most likely to be checked when haemoglobin is at least
context, hepcidin acts in an autocrine or paracrine manner to regulate 2 g/dl below the WHO threshold for anaemia2,3. Second, even when
the cellular LIP rather than systemic iron levels35–38. Hepatic hepcidin iron markers are investigated, often only ferritin is tested2. Even the
expression is controlled by bone morphogenetic protein 2 (BMP2) combination of ferritin and Tsat has limitations because these markers
and BMP6, signalling via a BMP receptor complex that involves several reflect the convergence of the multiple influences outlined above, they
accessory receptors, including homeostatic iron regulator protein, vary according to sex, age, ethnicity and smoking status, are subject to
haemojuvelin and TFR2 (ref. 91). Raised serum levels of hepcidin can diurnal variation, and are confounded by the presence of conditions
reflect increased hepatic expression due to high Tsat, inflammation such as haemoglobinopathy or blood loss through the gastrointestinal
(specifically IL-6 and IL-1β), liver injury and hyperlactataemia. Altera- tract or menstruation132–136. The lack of a clear, evidence-based defini-
tively, they can reflect impaired renal clearance of the hepcidin pep- tion of ID risks both undertreating patients who have a true unmet need
tide due to reduced glomerular filtration rate (GFR) such as in patients for iron and overdosing patients who do not need supplementary iron.
with kidney disease90,104–115. Conversely, reduced serum levels of hep- The past decade has seen efforts to reach a consensus definition of ID
cidin can be caused by hypoxia, low Tsat and the endocrine action of in the general population and in specific patient groups126–131 (Table 3).
erythroferrone, all of which suppress hepcidin expression90,91,104–115. In patients with HF, the diagnosis of ID is further confounded by
Erythroferrone is released by erythroblasts under the influence common comorbidities, such as diabetes mellitus, obesity, COPD
of erythropoietin108–110. Overstimulation of the erythropoietin– and chronic kidney disease (CKD), and certain medications such as
erythroferrone axis accounts for profound hepcidin suppression and β-blockers and sodium–glucose cotransporter 2 inhibitors137–142. Dapa-
results in non-transfusional iron overload in disorders of ineffective gliflozin, for example, was shown to reduce Tsat, ferritin and hepcidin
erythropoiesis108–110. Erythroferrone-mediated suppression of hepci- levels and to raise total iron-binding capacity and sTFR in patients with
din seems to overcome the stimulatory effects of IL-6, accounting for HF with reduced ejection fraction (HFrEF)142 through as-yet unknown
recovery from anaemia of inflammation (also known as anaemia of mechanisms. The ESC guidelines129 define ID as a ferritin concentration
chronic disease (ACD))30,111. Therefore, in the absence of inflammation, of <100 μg/l, or <300 μg/l with Tsat <20%, and they advocate testing
serum hepcidin levels reflect Tsat, hypoxia and erythropoietic activity. all patients with HF and supplementing those meeting this definition.
In inflammatory settings, hepcidin levels are increased (causing Tsat The rationale for this definition is to encompass both individuals with
to decline), but subsequent onset of anaemia can break the associa- absolute ID, in whom iron stores are truly depleted, and those with func-
tion between inflammation and hepcidin if erythroferrone levels are tional ID, in whom iron stores are not mobilizable due to elevated hep-
sufficiently raised. cidin levels. However, a prospective study published in 2022 indicated
that this definition does not adequately identify patients who are at
Erythropoietin. Erythropoietin influences iron homeostasis both by highest risk of adverse events or those most likely to benefit from
stimulating the erythropoietic demand for iron and by suppressing intravenous iron therapy137. Therefore, there is a disconnect between
hepcidin (via erythroferrone) to meet that demand108,116. Erythropoi- the current interpretation of iron markers and the true nature of the
etin is primarily produced by renal interstitial fibroblasts under the pathophysiology that they reflect or contribute to.
transcriptional control of hypoxia-inducible factor 2 (HIF2) in response
to hypoxia resulting from altitude, anaemia or pulmonary disease Relationship between iron status and HF
hypoxaemia117,118. Erythropoietin production is acutely upregulated Changes in iron markers during HF
by iron chelation because the degradation of HIF2 by prolyl hydroxy- Following acute cardiovascular events, the serum level of ferri-
lases requires iron as a cofactor119. Conversely, sustained ID blunts tin increases and that of transferrin decreases rapidly, as both are
erythropoietin production owing to translational repression of HIF2 by acute-phase proteins regulated by TNF42,85. Transient decreases in
IRP1 — a feedback mechanism that tethers erythropoietin production plasma iron levels and Tsat as well as a temporary elevation of the
to iron availability120. Indeed, mice lacking Irp1 and patients with IRP1 hepcidin level also occur after myocardial infarction, probably driven

Nature Reviews Cardiology


Review article

Diet: Fe2+
Haem: Fe2+
Fetus and Tablets: Fe2+
placenta
~500 mg Storage
~1.5 mg daily ~1,000 mg
Gut
Fe3+ Fe2+ 15

Fe3+ Fe2+ 14 Fe2+ Fe3+ Degradation 13

Mobilization as
per demand

18 16 17 11 Fe2+ Fe3+ 12 Ferritin (Fe3+)

10 Fe3+ 12

Uptake when
Tf(Fe3+)2 in excess
~3 mg
Non-erythroid pool
4 9 Degradation Ferritin (Fe3+) ~500 mg

As needed for 21
24 Fe 3+ 7
growth, renewal Mitochrondrion Sloughing
LIP Fe–S (Fe2+) ~1.5 mg daily
and function
NTBI Fe2+ 6 ETC
Intravenous Albumin:
iron Fe3+ Fe3+ 23 5 Haem (Fe2+) 8 Myoglobin (Fe2+)
Fe2+, Fe3+
~1,500 mg Citrate: Fe3+
Erythroid pool
Ferritin Ferritin Reticuloendothelial macrophages
16 17 ~600 mg
(?Fe3+) (?Fe3+)
~25 mg daily
Fe2+ HO-1
Fe3+ Fe2+ 2

22 Degradation

3
20
RBCs
~1,800 mg Transfusion
~200 mg
Free haem (Fe2+), Haemolysis
per unit
Cell-free haemoglobin
Haemoglobin (Fe2+)
(Fe2+) Menstruation
~1–50 mg per cycle
19

~25 mg daily Bone marrow


1 Fe3+ Fe2+ Haem (Fe2+) ~300 mg
Circulating pool

by IL-6 (refs. 85,86,143,144). Concomitantly, myocardial iron content low Tsat, and not low ferritin, are the strongest predictors of the risk
increases as a result of intramyocardial haemorrhage145. Therefore, of anaemia as HF progresses149. Furthermore, only a serum iron level of
caution must be used when inferring the iron status of a patient from ≤13 μmol/l or Tsat of ≤19.8% (not the ESC definition of ID) predict bone
snapshot measurements of iron markers in the hours after an acute marrow ID in patients with HFrEF149.
cardiovascular event. Progression towards anaemia stimulates erythropoietin pro-
In chronic HF (CHF), most of the available evidence derives from duction, which overcomes the inflammatory stimulus on hepcidin,
populations with ischaemic HF or HFrEF and shows that iron markers probably via erythroferrone. Indeed, hepcidin declines as patients
change gradually as disease progresses146–150 (Box 1). Hepcidin levels progress through NYHA classes, and this decline is strongly corre-
are elevated in the early stages of HF owing to the active inflammatory lated with the increase in sTFR level148. In advanced HF, the association
processes of ischaemic heart disease, infarct scarring and cardiac between hepcidin and IL-6 is broken, and hepcidin levels are lower than
remodelling144,148. Indeed, the association between hepcidin and IL-6 in earlier NYHA classes despite IL-6 levels being higher148. A gradual
is strongest in patients with NYHA class I HF148. An elevated hepcidin increase in erythropoietin level occurs over the course of HF pro-
level reduces serum iron availability, which begins to restrict erythro- gression, and its role in hepcidin suppression is supported by the
poiesis, eventually resulting in anaemia. Indeed, there is a progressive observation that plasma hepcidin levels are lower in patients with
reduction in serum iron and Tsat and a gradual increase in sTFR with CHF and anaemia than in healthy controls and correlate negatively
advancing NYHA class148. The role of iron restriction as a precursor to with erythropoietin levels147,150. Despite the suppression of hepcidin in
anaemia in CHF is supported by the finding that low serum iron and advanced HF, circulating iron availability continues to decline, which is

Nature Reviews Cardiology


Review article

Fig. 2 | The oxidative states, binding ligands and functional pools of taking up both Tf(Fe3+)2 via TFR-mediated endocytosis (10) and NTBI via NTBI
iron in the body. The various functional iron pools in the body are depicted transporters (11). This iron is primarily stored in ferritin cages (12); it can be
in different colours. Iron fluxes into and out of the body are shown in red released from ferritin (13) and exported via ferroportin into the circulation to
boxes around the edge. Most iron in the body is present in the erythroid pool meet extrahepatic demands (14). Iron taken up from the gut is also released into
(green area), either within reticuloendothelial macrophages, red blood cells the circulation via ferroportin (15). Iron released into the circulation as Fe2+ via
(RBCs) or the bone marrow. Haemoglobin synthesis in the bone marrow ferroportin at the sites of recycling, storage and absorption is oxidized to Fe3+
requires the uptake of ~25 mg of iron daily from the circulating pool (grey area) for loading onto transferrin (16). Some iron also joins the circulating NTBI pool,
in the form of transferrin (Fe3+) (1). This uptake is balanced by recycling of being bound to albumin or citrate (17). During pregnancy, the mother can lose
senescent RBCs by reticuloendothelial macrophages in the spleen, resulting up to 500 mg of iron to the fetus owing to the high expression of TFR on the
in ~25 mg of iron being released back into the circulation via ferroportin on the maternal side of the placenta (18). In women of reproductive age, up to 50 mg of
surface of macrophages (2). These macrophages also recycle iron from free iron is lost in each menstrual cycle (19). Every unit of blood transfusion delivers
haem or cell-free haemoglobin resulting from haemolysis (3). In non-erythroid 200 mg of iron in RBCs (20), which eventually joins the circulating iron pool
tissues (blue area), transferrin-bound iron Tf(Fe3+)2 is taken up by transferrin when transfused RBCs have been recycled. Some iron (~1.5 mg) is lost daily due
receptor (TFR)-mediated endocytosis (4), and some tissues additionally to sloughing of cells from the skin and mucosal surfaces (21) and is replenished
take up non-transferrin-bound iron (NTBI) via NTBI transporters (5). When by dietary iron uptake. Intravenous iron therapies can deliver up to 1,500 mg
inside the cells, iron joins the labile iron pool (LIP), before being used for of iron into the circulation. Most of this iron is enclosed in a carbohydrate shell
the synthesis of functional groups (6), such as those used in mitochondrial and is released into the circulation only following breakdown and ferroportin-
enzymes, or stored in ferritin (7). In cardiac and skeletal muscle, iron is also mediated export by reticuloendothelial macrophages (22). However, 10–40%
used for the synthesis of haem in myoglobin (8). Iron uptake via TFR-mediated of the iron is released directly into the circulation to join the NTBI pool (23) or
endocytosis can be increased (4) and iron from ferritin released (9) to meet loaded onto transferrin (24). Values shown are for an average, iron-replete adult.
cellular iron needs. Hepatocytes (yellow area) can buffer plasma iron levels by ETC, electron transport chain.

likely to reflect continued funnelling of iron towards the bone marrow rapid myocardial iron accumulation in the absence of any change in
(under the influence of erythropoietin) and, in some patients, truly serum iron availability35. Conversely, mice lacking cardiac hepcidin
depleted iron stores148. Notably, CKD (a common comorbidity) is an or harbouring a cardiac-specific substitution of ferroportin with a
important confounder of iron status in patients with HF owing to the hepcidin-resistant ferroportin develop myocardial iron depletion
impairment of erythropoietin production (renal anaemia) and reduced despite normal circulating iron levels36. Mice lacking cardiac TFR1
urinary clearance of hepcidin151. show early and fatal myocardial iron depletion172. These preclinical
studies also provide proof of concept that myocardial iron depletion
ID as a comorbidity in HF is sufficient to compromise the contractile function of the heart35,36,172.
Neither preclinical nor clinical studies have detected an increased risk However, studies examining the relationship between myocar-
of CVD in the setting of iron overload (such as in patients with thalas- dial iron levels and echocardiographic measures of heart function in
saemia or haemochromatosis), challenging the long-standing notion patients have yielded inconsistent results169,173,174. An important impli-
that iron is atherogenic152–155. By contrast, most prospective studies cation of the roles of hepcidin, ferroportin and TFR1 in myocardial
indicate that low serum iron availability, denoted specifically by low iron control is that myocardial iron depletion would be more likely
serum iron, Tsat or hepcidin (but not ferritin), predicts an increased risk in individuals with absolute ID (in whom both serum iron availability
of CVD events such as myocardial infarction and related adverse out- and hepcidin are reduced) than in patients with functional ID (in whom
comes, and worse long-term outcomes in patients with HF8,14,137,148,156–164 serum iron availability is reduced but hepcidin is elevated). Consistent
(Table 4). Several potential mechanisms underpin the role of ID in HF with this theory, in biopsy samples obtained during coronary artery
(Fig. 4), which are discussed in detail below. bypass graft (CABG) surgery, myocardial TFR1 expression (indicative of
an IRP-driven response to cellular ID) was higher in patients with abso-
Myocardial iron depletion. Whether myocardial iron depletion occurs lute ID (ferritin <100 μg/l) than in those with functional ID (Tsat <20%
in HF and whether it has disease-modifying effects remain subject to and ferritin 100–299 μg/l)171.
debate. Myocardial iron levels have been assessed in patients with HF Several mechanisms through which myocardial iron depletion
using magnetic resonance imaging (MRI) relaxometry parameters, could impair cardiac contractile function have been proposed such as
such as T1, R1 and T2*, and by direct quantification from myocardial a reduction in the activity of iron-dependent complexes in the electron
biopsy samples. However, MRI studies have either not detected an transport chain as well as a reduction in mitochondrial oxygen respira-
association between serum iron and myocardial iron levels or have tion and a shift from oxidative phosphorylation to glycolysis36,167–180.
detected only a trend towards reduced iron levels in myocardial and Impairments in respiration were shown to persist in vitro in isolated
skeletal muscle in patients with HF165,166. Taken together, myocardial myocardial mitochondria, suggesting an intrinsic decline in oxygen
biopsy studies support the notion that, in the early stages of HF, the utilization rather than simply reduced oxygen delivery175. By contrast,
myocardium maintains normal iron levels (despite reduced serum the only published study of myocardial energetics in living patients,
iron availability) by increasing the expression of TFR1 and decreasing using cardiovascular hyperpolarized MRI and phosphorus-31 magnetic
ferroportin locally. Subsequently, a decrease in TFR1 expression and resonance spectroscopy, showed that patients with non-ischaemic HF
an increase in ferroportin levels are associated with myocardial iron had preserved myocardial energetics176. As alluded to above, another
depletion with advancing HF167–171. proposed mechanism involves the upregulation of glycolysis in a man-
The importance of myocardial ferroportin and TFR1 for the main- ner that phenocopies HIF-driven responses, probably because iron
tenance of normal myocardial iron levels has been demonstrated in is a cofactor for the prolyl hydroxylases that regulate HIF stability,
preclinical models35,36. Mice lacking cardiomyocyte ferroportin show and indeed cellular ID can stabilize HIF and induce its transcriptional

Nature Reviews Cardiology


Review article

DMT1 Lactic acid


Duodenal
5 enterocyte 8
IL-6 Hepatocyte
O2 HIF Injury
1 6 HIF 10 O2

FPN
Ferroportin 9
Fe Hepcidin 4 Hepcidin 7

Erythrophagolysosome 14 TNF TFR


Fe
12 Transferrin 13 Fe Transferrin
Hb 21
Ferroportin Ferroportin
Fe 2 Fe Fe Fe 3
Ferritin
Transferrin
IRP 22 Ferritin 24 Ferritin 25 Injury
Kidney
23 Haemoglobin
Injury 11
TNF sTFR TNF
Splenic macrophage Consumption
27
Circulation Fe
18 15 Renal
EPO 19 HIF clearance
Transferrin O2
Senescence 26 IFNγ Fe Interstitial fibroblast
26
TFR 17

Senescence Haemoglobin IRP

Fe

Haemoglobin EPO 16 16 EPO Bone


marrow
20 RBC
Erythroblast Erythroblast
Gas exchange

Erythroferrone
Injury Lungs

Fig. 3 | Multifactorial influences on serum iron markers in health drive, which is stimulated by renal-derived erythropoietin (EPO) (16). Greater
and disease. Circulating markers of iron status are shown in the centre. erythroid demand is accompanied by greater erythroferrone production,
Arrows leading into the centre depict the tissue sources of each marker. aimed at suppressing hepcidin (9) and increasing iron availability in the
Regulatory influences in each tissue are also shown. Circulating iron levels are circulation. In erythroblasts, cellular iron demand for haemoglobin synthesis
dependent on ferroportin-mediated iron export from duodenal enterocytes is met via iron regulatory protein (IRP)-dependent upregulation of TFR (17).
(1), splenic reticuloendothelial macrophages (2) and hepatocytes (3). When haemoglobin synthesis is complete, TFR is shed into the circulation in
Ferroportin-mediated iron export from all three cell types is controlled by its soluble form (sTFR) (18). Therefore, sTFR levels reflect both the size of the
plasma hepcidin, which is primarily derived from hepatocytes (4). In duodenal erythroblast pool and its requirement for iron. Production of haemoglobin sets
enterocytes, iron absorption at the luminal side is dependent on several the oxygen-carrying capacity of the blood. Low haemoglobin production
transporters, including divalent metal transporter 1 (DMT1) (5). Iron absorption (anaemia) can result in hypoxaemia with consequences for iron absorption
is inhibited by injury (such as malabsorptive disorders) due to downregulation in the gut (1 and 5), hepcidin expression in hepatocytes (10) and erythropoietin
of DMT1 at the luminal side (5) and ferroportin at the apical side (1), and is production in renal interstitial fibroblasts (19). Impaired lung gas exchange
upregulated by hypoxia owing to the regulatory effects of hypoxia-inducible (for example, due to chronic obstructive pulmonary disease) can also result
factor (HIF) on duodenal DMT1 and ferroportin gene transcription. Hepcidin in hypoxaemia (20). Senescent red blood cells (RBCs) are engulfed by splenic
production in hepatocytes is upregulated by inflammation (IL-6) (6), macrophages (21), and recycled iron is subsequently released via ferroportin (2).
hepatocyte iron content (7) and anaerobic metabolism (lactic acid) (8), Inside these macrophages, ferritin expression is upregulated both by iron
and downregulated by the endocrine action of erythroferrone derived from availability (via IRP inhibition) (22) and by inflammation (via TNF) (23). Serum
the bone marrow (9) and the local action of hypoxia via HIF (10). Circulating ferritin derives from splenic macrophages (24), reflecting the interaction
hepcidin levels can also become elevated in chronic kidney disease due to between iron and inflammation in these cells. Serum ferritin levels can also be
impaired renal clearance (11). Serum transferrin is produced by hepatocytes augmented by leakage from hepatocytes due to liver injury (25). Inflammation
(12) in a manner that is downregulated by liver iron content (13) and can cause anaemia through iron-independent mechanisms. For example,
inflammation (tumour necrosis factor; TNF) (14). When in the circulation and IFNγ speeds up RBC senescence (26), whereas TNF and renal injury suppress
loaded with iron, transferrin is consumed via transferrin receptor (TFR)- erythropoietin production in interstitial fibroblasts (27). Iron-dependent
mediated endocytosis in tissues, most notably developing erythroblasts in mechanisms are also involved due to the elevation of hepcidin levels (8), which
the bone marrow (15). This rate of consumption is dependent on the erythroid reduces iron flux into the circulation.

Nature Reviews Cardiology


Review article

Table 2 | Factors that affect levels of circulating iron markers

Circulating Direction of Factors affecting change Mechanisms of change Refs.


iron marker change

Serum ferritin Increase Iron supplementation (intravenous or oral) Iron in macrophages inhibits IRP2, allowing translation of 23,43,44
ferritin heavy and light chains
Iron overload (for example, haemochromatosis, 23,43,44
iron-loading anaemias)
Inflammation (for example, acute cardiovascular events, TNF stimulates transcription of ferritin heavy chain in 66,68
chronic inflammation, infection) macrophages
Liver damage (for example, alcoholic or non-alcoholic Leakage from hepatocytes 64,65
liver disease)
Decrease Iron depletion (for example, absolute ID, iron chelation) Low iron in macrophages activates IRP2, inhibiting 23,43,44
translation of ferritin heavy and light chains
Serum Increase Depletion of liver stores (for example, pregnancy, Increased biosynthesis in the liver 42,76
transferrin absolute ID)
Hypoxia (for example, at altitude, hypoxaemia of Induction of transferrin gene expression by HIF1α 77
pulmonary disease)
Decrease Acute inflammation (for example, acute cardiovascular TNF inhibits transferrin biosynthesis in the liver and 84–87
events, infection) promotes transferrin uptake by macrophages
Repletion of liver iron stores (for example, intravenous Decreased biosynthesis in the liver 42,78
iron, haemochromatosis, thalassaemia)
Liver damage (for example, alcoholic or non-alcoholic 79,80
liver disease)
Erythropoietic expansion (for example, response Expansion of the erythroid pool accompanied by increased 42,116
to erythropoietin, ineffective erythropoiesis (as in transferrin uptake via TFR1 on erythroblasts
β-thalassaemia) and erythroid anaemia due to vitamin
B12 deficiency)
Nephrosis Excess renal clearance 42,81,82
Serum iron Increase Iron supplementation (intravenous or oral) Increased iron release from gut enterocytes (oral iron), 54,55,
direct iron release into the circulation (intravenous iron) 57,58
Hypoxia (for example, at altitude, hypoxaemia of HIF stimulates expression of DMT1 for iron uptake at the 92,93
pulmonary disease) lumen side and ferroportin for iron export at the basolateral
side of gut enterocytes
Liver injury (for example, alcoholic or non-alcoholic liver Decreased serum transferrin increases NTBI 79,80
disease)
Decrease Inflammation (for example, acute cardiovascular events, Increased iron uptake into macrophages via DMT1; increased 46,90,91
infection, chronic disease) levels of endogenous chelators (for example, lactoferrins and
lipocalin 2); decreased levels of ferroportin in gut enterocytes
and splenic macrophages due to hepcidin elevation
Erythropoietic expansion (for example, response to Expansion of the erythroid pool increases transferrin (Fe3+) 27,116
erythropoietin) turnover
Increase or Time of day (a.m. = increase; p.m. = decrease) Reflects variation in hepcidin level 88
decrease
Transferrin Increase Acute inflammation (for example, acute cardiovascular Decreased serum transferrin 83–85
saturation events, infection)
Liver injury (for example, alcoholic or non-alcoholic liver 79,80
disease)
Iron supplementation (intravenous or oral) Increased serum iron 54,55,
57,58
Decrease Inflammation (for example, chronic disease, infection) Decreased serum iron 46,90,91
Iron chelation, phlebotomy 212
Increase or Time of day (a.m. = increase; p.m. = decrease) Reflects variation in serum iron level 87
decrease
sTFR Increase Unmet erythroid iron demand (for example, ID) Reduced LIP in erythroblasts activates IRP to increase surface 97
expression of TFR1, which is subsequently shed as sTFR
Erythropoietic expansion (for example, erythropoietin Expansion of erythroblast pool leads to more TFR1 being 98,99
response, haemolytic or sickle cell anaemia, ineffective shed into the circulation as sTFR
erythropoiesis)

Nature Reviews Cardiology


Review article

Table 2 (continued) | Factors that affect levels of circulating iron markers

Circulating Direction of Factors affecting change Mechanisms of change Refs.


iron marker change

sTFR Decrease Suppressed erythropoiesis (for example, following Contraction of the erythroblast pool leads to less TFR1 being 100,101
(continued) chemotherapy, impaired erythropoietin production in shed into the circulation as sTFR
renal disease)
Hepcidin Increase Iron supplementation (intravenous or oral) Tsat elevation stimulates hepcidin transcription by 36,57
increasing BMPR2 signalling on hepatocytes
Inflammation (for example, chronic disease, infection, IL-6 signals at IL-6 receptor on hepatocytes to stimulate 90,91,
acute cardiovascular events) hepcidin transcription via a STAT3 binding site in hepcidin 105,111
promoter; IL-1β increases C/EBPδ expression in hepatocytes,
which activates hepcidin transcription
Liver injury (for example, alcoholic or non-alcoholic Endoplasmic reticulum stress in hepatocytes activates 106
liver disease) DDIT3, which increases C/EBPα activity and hepcidin
transcription
Hyperlactataemia (for example, in athletes, Lactate interaction with soluble adenylyl cyclase raises 114
some cancers) cAMP levels, which activates SMAD1/5/8 signalling and
increases hepcidin transcription in hepatocytes
Renal disease with reduced eGFR Impaired hepcidin clearance from circulation 115
Decrease Erythropoietic expansion (for example, anaemia, Differentiating erythroblasts release the hormone 30,108–111
response to erythropoietin, ineffective erythropoiesis erythroferrone, which inhibits BMPR2 signalling in
(as in β-thalassaemia) and erythroid anaemia due to hepatocytes to suppress hepcidin transcription
vitamin B12 deficiency)
Hypoxia (for example, at altitude, hypoxaemia of HIF stimulates the expression of furin and matriptase 2, 112,113
pulmonary disease) proteases that cleave the BMP co-receptor haemojuvelin
from the surface of hepatocytes, reducing hepcidin
transcription
Erythropoietin Increase Anaemia Reduced oxygen delivery to interstitial fibroblasts inhibits 108,117,118
HIF prolyl hydroxylases; HIF2 is stabilized and drives
transcription of the erythropoietin gene
Hypoxia (for example, at altitude, hypoxaemia of Local hypoxia in interstitial fibroblast inhibits HIF prolyl 117,121
pulmonary disease) hydroxylases; HIF2 is stabilized and drives transcription of
the erythropoietin gene
Iron chelation Removal of intracellular iron from interstitial fibroblast 119
inhibits HIF prolyl hydroxylases (they require iron as a
cofactor); HIF2 is stabilized and drives transcription of the
erythropoietin gene
Decrease Iron deficiency with low Tsat Reduced iron delivery to interstitial fibroblasts activates 119–121
IRP1, which causes translational inhibition of HIF2, thereby
reducing erythropoietin transcription
Inflammation (for example, chronic disease, infection, TNF decreases renal erythropoietin production 124,125
acute cardiovascular events)
BMP, bone morphogenetic protein; BMPR2, bone morphogenetic protein receptor type 2; C/EBP, CCAAT/enhancer-binding protein; DDIT3, DNA damage-inducible transcript 3 protein
(also known as CAAT/enhancer-binding protein homologous protein, CHOP); DMT1, divalent metal transporter 1; eGFR, estimated glomerular filtration rate; HIF, hypoxia-inducible factor;
ID, iron deficiency; IRP, iron regulatory protein; LIP, labile iron pool; NTBI, non-transferrin-bound iron; SMAD, mothers against decapentaplegic homologue; STAT3, signal transducer and
activator of transcription 3; sTFR, soluble transferrin receptor; TFR, transferrin receptor; TNF, tumour necrosis factor; Tsat, transferrin saturation.

targets, even in normoxia61,179. Disruption of calcium signalling genes In COPD, ID and low circulating hepcidin levels are associated with
has also been proposed, involving the downregulation of ryanodine increased skeletal muscle IRPs (consistent with intracellular ID) and
receptor 2 and a reduction in sarcoplasmic/endoplasmic reticulum Ca2+ with reduced exercise capacity186. The effects of ID on skeletal muscle
ATPase activity in the myocardium180. Impaired oxygen storage due to function and physical activity have long been recognized and are not
reduced myoglobin synthesis has also been suggested as a potential unique to patients with HF26,187–189.
mechanism, although this phenomenon has been observed only in
skeletal muscle and not in the myocardium181,182. Pulmonary disease. COPD is one of the major comorbidities in HF,
affecting 10–40% of patients190,191. ID is common in COPD and is para-
Skeletal muscle iron depletion. Exercise capacity is often reduced doxically accompanied by iron accumulation in lung parenchyma,
in patients with HF and universally predicts worse outcomes173,183. In a phenomenon that has been confirmed in preclinical models192–194.
HF, ID is also associated with reduced exercise capacity and increased Lung epithelial cells and alveolar macrophages express ferroportin, and
muscle wasting184,185. The observation that ID reduces myoglobin in skel- elevated hepcidin levels seen in advanced COPD have been suggested
etal muscle provides a potential mechanism for this association181,182. to promote both serum ID and iron accumulation in the lungs, which

Nature Reviews Cardiology


Review article

is an important driver of fibrosis and hypoxaemia194,195. Patients with (PharmaNutra), have been suggested to have greater fractional absorp-
COPD and ID were found to be more hypoxaemic than their iron-replete tion potentially because the slow release of iron blunts the subsequent
counterparts and intravenous iron therapy improved exercise capacity rise in hepcidin218–221. Fractional absorption is enhanced by alternate-day
without improving hypoxaemia192,196. Therefore, in patients with HF dosing, which avoids sustained hepcidin elevation, and by dosing in
and COPD, the presence of functional ID could contribute to worse the early morning or in the evening when hepcidin levels are low222–224.
HF outcomes by compounding iron accumulation in lung parenchyma, Malabsorptive disorders, such as coeliac disease and Crohn’s disease,
worsening fibrosis and hypoxaemia. Furthermore, interpretation of reduce fractional absorption, rendering patients refractory to oral iron
iron markers in patients with HF and severe COPD could be confounded therapy225,226. The notion that inflammation blocks the absorption of
by the presence of hypoxaemia-driven polycythaemia141,197,198. iron from oral iron supplementation by raising hepcidin levels provided
the rationale for the move towards intravenous iron therapies. However,
Haemodynamic stress on the right heart. Pulmonary hyperten- some evidence exists that this paradigm might not apply to individuals
sion affects an estimated 36–83% of patients with HFpEF and is also with ID anaemia. Indeed, influenza and diphtheria–pertussis–tetanus
a frequent complication of COPD199–202. Pulmonary hypertension is vaccinations were shown to raise hepcidin levels and decrease frac-
characterized by remodelling of the pulmonary circulation, leading to tional iron absorption from a 57Fe-labelled test meal in women without
chronically raised pulmonary arterial pressure (PAP), haemodynamic anaemia but the vaccines had no effects on women with ID anaemia111.
stress on the right ventricle and ultimately right HF203. ID is common These findings echo previous observations that anaemia breaks the
in pulmonary hypertension and is associated with increased PAP and association between inflammation and hepcidin30,109.
NYHA functional class, poor survival, and elevated markers of car- In patients with HF, oral iron therapy often improves haemato-
diovascular dysfunction204–208. In healthy individuals, the pulmonary logical and circulating iron parameters but its effects on functional
vascular response to hypoxia is potentiated by clinical ID or pharma- and clinical outcomes have not been consistent227–234. The conflict-
cological iron chelation and is attenuated by repleted iron stores or ing findings are likely to reflect differences between studies in the
iron supplementation209–213. The speed by which iron chelation or sup- total dose of iron delivered to the circulation that, in turn, depends
plementation modifies the pulmonary vascular response to hypoxia on whether the formulation has fast or slow release, the frequency of
indicates a haemoglobin-independent mechanism, probably relating to dosing, and the presence and severity of inflammation and anaemia30,111.
iron levels in pulmonary vascular cells212,213. Consistent with this theory, Oral iron therapy seems to be less effective for preventing or treat-
preclinical studies have shown that ID in pulmonary arterial smooth ing acute blood loss anaemia, for example, in the setting of CABG
muscle cells is sufficient to cause pulmonary hypertension and right surgery234,235, than for chronic anaemia.
HF by stimulating the production of the vasoconstrictor endothelin 1 One underexplored question is the extent to which oral iron therapy
in a manner that could be prevented, yet only partially reversed, influences myocardial iron levels. In a mouse model of iron-refractory
by intravenous iron therapy38. ID anaemia (harbouring a deletion of the Tmprss6 gene, which encodes
In patients with pulmonary vascular disease, the definition of transmembrane serine protease 6, causing excessive hepcidin produc-
ID as Tsat <21% (but not the ESC definition) identified patients with tion), administration of sucrosomial iron corrected ID and restored
reduced peak oxygen consumption (VO2) or worse performance in liver iron stores but did not increase myocardial iron levels220. By
the 6-min walking test207. Patients with ID had similar left ventricular contrast, in neonatal piglets with anaemia, oral administration of
function and structure to those without but had more right ventricular ferrous glycine chelate increased myocardial iron levels by ~50%236.
remodelling, including larger right ventricular volume and mass and
lower right ventricular ejection fraction207. Trials of iron supplementa-
tion in pulmonary hypertension have not shown reproducible effects Table 3 | Current definitions of iron deficiency
on functional parameters despite improvements in haematological
parameters214. These findings potentially reflect a level of pulmonary Population Ferritin Transferrin Refs.
(µg/l) saturation (%)
vascular remodelling that is irreversible as seen in preclinical models38.
Therefore, absolute ID can promote or exacerbate pulmonary hyper- Healthy individuals
tension, and these effects could contribute to worsening outcomes in Adults (aged >20 years) <15 No cut-off 126
patients with HFpEF or COPD.
Children and adolescents (aged 5–20 years) <15 No cut-off 126

Iron supplementation in HF Infants (aged 0–5 years) <12 No cut-off 126


The balance between the benefits and harms of iron supplementation Pregnant women <15 <16 126,127
depends on whether therapies are given to individuals with a truly
Individuals with infection or inflammation
unmet need for iron and whether they meet that need in a timely and
safe manner. In this section, we examine findings from key clinical trials Adults (aged >20 years) <70 No cut-off 126

of oral and intravenous iron therapies in patients with HF (Table 5). Children and adolescents (aged 5–20 years) <70 No cut-off 126
Infants (aged 0–5 years) <30 No cut-off 126
Oral iron therapy
Individuals with kidney disease
Oral formulations differ according to the complexing ligand and the
amount of elemental iron they contain. However, these agents typically All individuals <500 <30 128
deliver ~100 mg of ferrous iron into the gut lumen, where uptake is Individuals with heart failure
dependent on DMT1, and fractional absorption of iron to the basolateral All individuals <100 No cut-off 129
side is 10–22%21,215,216. Tsat and serum hepcidin subsequently rise within
<300 <20
4 h and 8 h, respectively54,217. Novel formulations, such sucrosomial iron

Nature Reviews Cardiology


Review article

Box 1

Evolution of serum iron markers in chronic HF


The model is based on observational and longitudinal cohort studies stimulation to IRP-mediated translational repression of the ferritin
of patients with chronic heart failure (HF). The mechanisms at play transcript in macrophages.
and the levels of serum iron markers change with disease progression
(see the figure). NYHA class IV
Worsening disease and comorbidities heighten inflammation, but
NYHA class I hepcidin is now dominantly suppressed by bone marrow-derived
Adverse cardiac remodelling triggers inflammation (IL-6), which erythroferrone and low Tsat. These circumstances enable iron to
increases hepcidin production. Serum ferritin levels are raised, be mobilized from the macrophages, resulting in IRP-mediated
reflecting tumour necrosis factor (TNF) signalling and translational translational repression of ferritin and a further drop in serum
de-repression of the ferritin transcript iron regulatory proteins (IRPs) ferritin level. Despite the low hepcidin concentration, Tsat
(caused by iron retention in macrophages). and serum iron remain low due to increased iron utilization by
the bone marrow, which is required to recover haemoglobin
NYHA class II synthesis.
Adverse cardiac remodelling and worsening comorbidities sustain
inflammation and continue to stimulate hepcidin production.
However, a hepcidin-mediated drop in transferrin saturation
(Tsat) begins to exert negative feedback on hepcidin expression.
Serum iron marker level

Consequently, iron retention in macrophages is partially relieved,


activating IRPs and causing translational repression of ferritin,
resulting in a relative decrease in serum ferritin levels.

NYHA class III


The now sustained lowering of Tsat restricts erythropoiesis
(as indicated by an increase in the soluble transferrin receptor I II III IV
(sTFR) level), and the haemoglobin level begins to fall. Anaemia
Worsening HF (NYHA class)
stimulates the synthesis of erythropoietin and erythropoiesis in the
bone marrow, thus releasing erythroferrone. This hormone exerts Heathy levels IL-6 and CRP Haemoglobin
sTFR Ferritin
a dominant inhibitory effect on hepcidin, overcoming the IL-6 Hepcidin Serum iron and Tsat
signal. The serum ferritin level drops, reflecting the shift from TNF

An important difference between the mouse and piglet studies is that decade, the percentage of patients with HF and ID receiving intravenous
hepcidin was elevated in the mouse model (owing to the loss of trans- iron has grown to 19% in Sweden, 33% in Spain, 39% in France and 49% in
membrane serine protease 6) and was probably suppressed in the piglet the UK232–242. Several trials have shown that intravenous iron administra-
model due to the dominant inhibitory effect of anaemia. This factor tion reduces the risk of recurrent hospitalization for HF or cardiovas-
raises the intriguing possibility that hepcidin not only modifies the cular death and/or improves functional capacity (typically measured
fractional absorption of oral iron but also its subsequent retention in by the 6-min walking test or peak VO2)243–255. However, some of these
ferroportin-expressing tissues such as the myocardium. benefits were not reproduced in other trials. In the placebo-controlled
HEART-FID trial255 of intravenous iron therapy (FCM) in patients with
Intravenous iron therapy HFrEF, a significance level of P = 0.01 was set to enable an assessment of
Clinically approved intravenous iron formulations deliver iron inside efficacy based on a single randomized trial. No sufficiently significant
a carbohydrate shell — disaccharide in iron sucrose, branched poly- improvements were identified in the hierarchical composite outcome
saccharide in iron dextran and ferric carboxymaltose (FCM), or linear of death, hospitalization for HF or change in 6-min walking distance
oligosaccharide in ferric derisomaltose (also known as iron isomalto- with the use of intravenous iron (P = 0.02)255. The IRONMAN study252
side)237. Unlike oral iron, which delivers incremental increases in serum of iron isomaltoside in patients with HFrEF did not identify signifi-
iron levels, a single bolus dose of intravenous iron can deliver up to cant improvements in functional capacity despite reducing the risk of
1,500 mg of iron directly into the circulation. For example, FCM infu- hospital admissions for HF and cardiovascular death.
sion increases serum iron levels within 30 min, with Tsat subsequently The divergent findings from these trials are likely to reflect the
exceeding 90%57. Some intravenous iron formulations release iron in collective effects of differences in the definition of ID, minimum hae-
two stages: first rapidly and directly into the circulation as NTBI and moglobin concentration cut-off, the type and dose of iron formula-
then indirectly following breakdown and release by macrophages57,58. tion, and the HF phenotype of the participants. In most trials, the ESC
The ESC guidelines advocate the use of intravenous iron therapy definition of ID was used and, therefore, probably captured a mix
in all patients with HF who meet its definition of ID129. Over the past of patients with functional or absolute ID. In the IRONMAN trial252,

Nature Reviews Cardiology


Review article

a ferritin cut-off of 400 µg/l was used and probably captured a greater hyperpolarized MRI and phosphorus-31 magnetic resonance spectros-
proportion of patients with functional ID, whereas Toblli et al. defined copy should help to determine the effects of intravenous iron therapy
ID as Tsat <20% and ferritin <100 ng/ml, which probably enriched on myocardial energetics, including whether these effects manifest
their study with patients who had absolute ID243,252. The type of ID is before or after improvement in haemoglobin concentration.
relevant because it can influence HF prognosis. Indeed, low Tsat alone
or the combination of anaemia with high ferritin have been shown to Improved skeletal muscle energetics. Among patients with chronic
predict poor prognosis in HF256,257. Trials have also used various intra- HFrEF (NYHA class ≥II) and ID, iron isomaltoside versus placebo was
venous iron formulations known to have differing pharmacokinetic found to improve phosphocreatine and ADP recovery half-times, rest-
properties. For instance, NTBI is low with iron isomaltoside, moderate ing respiratory rate and post-exercise Borg dyspnoea score at 12 weeks
and more acute with iron sucrose, and high and more sustained with after infusion264. FCM was also found to improve peak VO2 in a similar
FCM. This variation results in an area under the curve for NTBI that is timeframe, which was related to the extent of change in haemoglobin
sevenfold greater for FCM than for the other two formulations57,58. The level, and skeletal muscle iron content did not change166. Together,
distinct pharmacokinetics of these iron formulations have been dem- these data suggest that the improvement in exercise capacity might
onstrated in otherwise healthy volunteers with anaemia (absolute ID) be driven by improved oxygen delivery rather than by iron repletion
but have yet to be compared in the contexts of HF or functional ID57,58. to the skeletal muscle.
A minimum haemoglobin concentration cut-off (typically 8–9 g/dl)
was set in some trials but not others, meaning that the proportions Improved respiratory and ventilatory functions. In patients with HFrEF,
of patients with moderate or severe anaemia varied between studies. anaemia and ID, FCM improved the hypercapnic ventilatory response
These patients with severe anaemia typically have worse prognoses but and sleep-disordered breathing within 2 weeks, with subsequent
derive greater benefit from haemoglobin correction than those with
mild anaemia. The HF phenotype of participants also differed between
trials according to left ventricular ejection fraction threshold, use of Table 4 | Cardiovascular risk and related adverse outcomes
plasma markers and NHYA functional class243–255. Although seemingly associated with ID
subtle, these variations would have resulted in different proportions
of patients with HFrEF and those with HFpEF, with correspondingly Population Findings Ref.
distinct aetiologies and comorbidities. Risk of cardiovascular events
Overall, the body of evidence indicates that some but not all
Oldera individuals Low serum iron predicts an increased risk of 156
patients with HF derive benefits from intravenous iron therapy. first CVD in women only
Responder analyses have identified low Tsat, diabetes mellitus,
Low serum iron predicts an increased risk of 158
impaired renal function, ischaemic heart disease and anaemia as pre- acute MI and stroke
dictors of those who will derive the greatest benefits from intravenous
Individuals with high Low serum iron (but not Tsat or TIBC) predicts 159
iron therapy in terms of reducing recurrent hospitalization from HF and
cardiovascular risk an increased risk of MI in women only
cardiovascular death254. In patients who had undergone heart trans-
plantation within 1 year before study enrolment, those with ferritin Healthy individuals Lowest quintiles for serum iron and Tsat 160
predict highest risk of MI in the short term
<30 μg/l or sTFR above the reference value had the greatest improve-
ment in peak oxygen uptake following intravenous iron therapy, and Mixedb Serum iron and Tsat inversely related to the 161
risk of CHD; haem iron positively related to
this group also had significantly reduced hepcidin levels, consistent the risk of CHD; ferritin shows borderline
with absolute ID258,259. One interpretation of responder analyses is that positive association with the risk of CHD
different patient subgroups derive benefits through different path-
Adverse outcomes after cardiovascular events
ways. For example, among patients with anaemia, the benefit is likely
to be mediated through haemoglobin correction, whereas in patients Patients discharged Absolute ID but not functional ID predicts an 9
from hospital after AHF increased risk of 30-day hospital readmission
without anaemia, fulfilment of an unmet need outside the erythron is
the probable mechanism. Conversely, the lack of a response to intra- Patients with STEMI Low serum iron predicts the risk of 14
developing HF regardless of ferritin or
venous iron therapy in some patients suggests that their diagnosis of haemoglobin
ID does not reflect an unmet need for iron or that the unmet need is
Patients with AHF Combination of low hepcidin and high sTFR 163
not being adequately fulfilled by intravenous iron therapy. Future tri-
(excluding ACS) predicts the risk of death
als need to be sufficiently powered to resolve differences in response
between absolute and functional ID as well as the modifying effects of Healthy individuals Lowest quartile for Tsat or serum iron 164
predicts the highest risk of death from IHD
anaemia, inflammation and HF phenotype.
Adverse outcomes in patients with CHF
Mechanisms of benefit for intravenous iron Patients with HF Tsat <20% and serum iron ≤13 μmol/l predict 137
Improved myocardial energetics. Improvement in echocardiographic increased all-cause mortality over 5 years
parameters of heart function has been reported in some trials of intra- Patients with systolic HF Low Tsat and low hepcidin (but not ferritin) 148
venous iron therapy in HF260–262. In preclinical models, intravenous iron predict increased risk of 3-year mortality
therapy prevented the energetic impairment and contractile dysfunc- ACS, acute coronary syndrome; AHF, acute heart failure; CHD, coronary heart disease;
tion caused by global or myocardial ID36,170. Improvements in energetics CHF, chronic heart failure; CVD, cardiovascular disease; HF, heart failure; ID, iron deficiency;
IHD, ischaemic heart disease; MI, myocardial infarction; STEMI, ST-segment elevation
could be the result of either increased oxygen delivery to the myocar-
myocardial infarction; sTRF, soluble transferrin receptor; TIBC, total iron-binding capacity;
dium due to improved haemoglobin concentration or increased oxygen Tsat, transferrinsaturation. aMen aged 55–80 years, women aged 60–80 years. bMeta-analysis
utilization following myocardial iron repletion165,166,263. Cardiovascular of 21 prospective cohort studies.

Nature Reviews Cardiology


Review article

Absolute ID Reduced serum Functional ID


• Depleted iron stores iron availability • Inflammation
• Increased iron demand (chronic disease,
• Blood loss infection)

Bone Kidney Lung epithelium


marrow
Iron accumulation
↓ Cellular iron uptake ↓ Haemoglobin ↓ Erythropoietin

Fibrosis

Impaired gas
exchange

↓ Oxygen delivery

Cellular iron Cellular hypoxia


depletion

Chronic activation Pulmonary


Endothelin 1 vasculature
of HIF signalling

Skeletal muscle Myocardium Pulmonary


Impaired ETC activity Impaired Glycolytic Impaired Ca2+ hypertension
myoglobin metabolism signalling
synthesis
Reduced oxidative
phosphorylation
Energetic
insufficiency

Reduced exercise Reduced cardiac ↑ Haemodynamic stress


capacity contractility

↑ HF hospitalization and mortality

Fig. 4 | Potential mechanisms linking ID with poor outcomes in HF. Interconnections between functional and absolute iron deficiency (ID), and the mechanisms
involving haemoglobin-dependent and haemoglobin-independent pathways. ETC, electron transport chain; HF, heart failure; HIF, hypoxia-inducible factor.

improvements in oxygen delivery to muscles and patient-reported in a subgroup analysis of the FAIR-HF trial245,246, FCM improved eGFR
outcomes, particularly fatigue, when compared with placebo265. after week 24. Therefore, in some patients, improved HF outcomes
In iron-deficient but otherwise healthy volunteers, FCM reduces PAP might relate to improved renal function. Given the high prevalence
and blunts the pulmonary vascular responses to hypoxia associated of CKD in patients with HF, more studies are needed to specifically
with inhibition of the vasoconstrictor endothelin 1 (refs. 38,213). There- examine the interaction between CKD and HF in terms of intravenous
fore, some patients with HF (such as those with pulmonary hyperten- iron therapy outcomes.
sion or COPD) might derive benefits from intravenous iron therapy via
improved respiratory and ventilatory functions. Unresolved questions
The fate of intravenous iron. Intravenous therapies deliver a supra-
Decongestion. Fluid congestion is common in CHF and is an independ- physiological dose of iron rapidly and directly into the circulation.
ent predictor of prognosis. In the placebo-controlled FAIR-HF trial266 In healthy volunteers, MRI T1 values of the myocardium, liver, spleen
of 459 patients with CHF and ID, FCM reduced body weight and plasma and skeletal muscle were all shortened (indicating a greater tissue
volume status (a novel index of congestion), which was associated with concentration of iron) within 60 min of FCM infusion267. In rats, the
improvements in 6-min walking distance and NYHA class. Plausible MRI R2 relaxometry parameter increased (indicating a greater tissue
mechanisms through which iron supplementation reduces plasma concentration of iron) in the liver, spleen and kidneys within 10–20 min
volume status include improved renal function or a reduction in the of iron sucrose infusion, only decreasing over 24 h in the kidneys,
level of catecholamines that normally activate the pro-congestive probably reflecting renal iron reabsorption268. Importantly, there
renin–angiotensin–aldosterone pathway. were no changes in bone marrow or muscle R2 signals over this time
course268. The rapid increase in iron levels in some non-erythroid tis-
Improved renal function. In some trials of intravenous iron therapy, sues suggests direct circulatory uptake of the NTBI released from the
up to 40% of patients with HF had an estimated GFR (eGFR) consistent carbohydrate shell57,58. Evidence for the physiological consequences of
with stage 3a CKD245,247. Patients with low eGFR were found to derive this process exists; in healthy volunteers, the hypoxia-induced rise in
greater benefits from intravenous iron therapy in terms of change in PAP was blunted within 60 min of FCM infusion before any detectable
6-min walking distance than those with normal eGFR. Furthermore, rise in haemoglobin level213.

Nature Reviews Cardiology


Review article

Table 5 | Effects of oral and intravenous iron therapies in HF

Study Formulation and dose Population Follow-up Parameters improved Parameters not Ref.
duration improved

Oral iron

Crosby et al. Elemental iron 50 mg or Patients undergoing CABG surgery 59 days None Red blood cell 227
200 mg daily for 2 months mass, ferritin
vs placebo

Clevenger Various oral iron formulations Adults with anaemia and without 1 yeara ∆Haemoglobin (0.91 g/dl vs Mortality 229
et al. (meta-analysisa) CKD, including a subgroup with HF 1.04 g/dl (for parenteral iron));
requirement for blood transfusion
(RR 0.66 vs RR 0.84 (for parenteral
iron)) compared with inactive
control

IRONOUT Oral iron polysaccharide HFrEF with ID (ferritin 15–100 µg/l 16 weeks ∆Serum iron (effect of treatment Exercise 230
150 mg twice daily for 16 weeks or ferritin of 101–299 µg/l with 11 µg/dl); ∆Tsat (effect of capacity
vs placebo Tsat <20%) treatment +3.3%); ∆sTFR
(effect of treatment –0.3 mg/ml)

Zdravkovic Ferrous fumarate 115 mg twice Chronic DHF with ID (ferritin 6 months ∆Haemoglobin (10.45 g/l vs None 232
et al. daily for 6 months or ferric <100 µg/l with Tsat <20%) 4.56 g/l); ∆Tsat (21.0% vs 23.4%);
hydroxide polymaltose 100 mg ∆6MWD (62.07 m vs 35.38 m);
twice daily for 6 months NYHA class

Karavidas et al. Sucrosomial iron 28 mg daily HFrEF with ID (ferritin <100 ng/ml 3 months ∆Haemoglobin (0.37 ± 0.55 g/dl None 233
for 3 months vs no treatment or ferritin 100–299 ng/ml with Tsat vs –0.21 ± 0.47 g/dl); ∆serum
<20%) iron (17.58 ± 11.73 mg/dl vs
−1.36 ± 8.68 mg/dl); ∆ferritin
(28.34 ± 31.39 ng/ml vs
−0.05 ± 6.97 ng/ml); ∆6MWD
(9.50 ± 15.83 m vs −3.00 ± 8.81 m);
∆KCCQ (4 ± 5 vs –1 ± 3)

Suryani et al. Oral ferrous sulfate, 200 mg Patients with HFrEF and ID anaemia 12 weeks ∆Haemoglobin (1.0 g/dl vs –0.1 g/dl); None 234
three times per day for (ferritin <100 µg/l or 100–300 µg/l ∆ferritin (86 ng/ml vs 2 ng/ml);
12 weeks vs placebo with Tsat <20%) ∆Tsat (12.4% vs 3.0%), ∆6MWD
(46.23 ± 35.00 m vs –13.70 ± 46.00 m)

Oral vs intravenous iron

IRON-HF Intravenous iron sucrose Patients with HF and anaemia 3 months ∆Haemoglobin (1.04 g/dl oral Peak VO2 228
200 mg once per week for (ferritin <500 µg/l with Tsat <20%) vs 1.69 g/dl intravenous iron (oral iron)
5 weeks or oral ferrous sulfate vs 1.1 g/dl placebo); ∆ferritin
200 mg three times per day for (126 µg/l oral vs 103 µg/l intravenous
8 weeks vs placebo iron vs –42 µg/l placebo); ∆peak
VO2 (3.5 ml/kg/min; intravenous
iron only)

Garrido-Martin Intravenous iron(III)-hydroxide Patients undergoing CABG surgery 1 month Ferritin levels at hospital discharge Anaemia status, 235
et al. sucrose complex three (1,321 ± 495 ng/ml vs 541 ± 471 ng/ml transfusion
doses of 100 mg per day (intravenous iron only)) requirement
during preoperative and
postoperative hospitalization
or oral ferrous fumarate
iron 105 mg (one pill) per
day preoperatively and
postoperatively and for
1 month after hospital
discharge

Intravenous iron

Toblli et al. Iron sucrose 200 mg weekly for LVEF ≤35%, Tsat <20%, ferritin 6 months LVEF (35.7 ± 4.7% vs 28.8 ± 2.4%); None 243
5 weeks vs placebo <100 µg/l NYHA functional class (2.0 ± 0.2
vs 3.3 ± 0.6); 6MWD (240.1 ± 51.2 m
vs 184.5 ± 58.5 m); MLHFQ score
(41 ± 7 vs 59 ± 8)

FERRIC-HF Iron sucrose 200 mg weekly HF NYHA class II/III, LVEF ≤45%, 16 weeks Ferritin (treatment effect 273 ng/ml); Haemoglobin 244
until ferritin >500 µg/l, 200 mg ferritin <100 μg/l or 100–300 μg/l peak VO2 (treatment effect
monthly thereafter for 16 weeks with Tsat <20% 2.2 ml/kg/min); NYHA functional
vs no treatment class (treatment effect –0.6)

Nature Reviews Cardiology


Review article

Table 5 (continued) | Effects of oral and intravenous iron therapies in HF

Study Formulation and dose Population Follow-up Parameters improved Parameters not Ref.
duration improved

Intravenous iron (continued)


FAIR-HF Ferric carboxymaltose weekly CHF (NYHA class II/III), LVEF 24 weeks Haemoglobin (130 ± 1 g/l vs None 245
until repletion, then every ≤40%; minimum haemoglobin 125 ± 1 g/l); ferritin (323 ± 13 μg/l vs
2 weeks during maintenance cut-off 9.5 g/dl 74 ± 8 μg/l); Tsat (29 ± 1% vs 19 ± 1%);
phase vs placebo 6MWD (treatment effect 35 ± 8 m)
and quality-of-life assessments
(KCCQ and EQ-5D: OR 2.51 for
improvement)
CONFIRM-HF Ferric carboxymaltose CHF (NYHA class II/III), LVEF ≤45% 52 weeks ∆Haemoglobin (treatment effect None 247
2–5 doses over 36 weeks and elevated natriuretic peptide 1.0 ± 0.2 g/dl); ∆ferritin (treatment
vs placebo levels effect 200 ± 19 ng/ml); ∆Tsat
(treatment effect 5.7 ± 1.2%);
risk of recurrent hospitalization for
HF (HR 0.71); ∆6MWD (treatment
effect 36 m)
EFFECT-HF Ferric carboxymaltose up HF (NYHA class II/III), LVEF ≤45%, 24 weeks Haemoglobin (13.9 ± 1.3 g/dl vs None 248
to 3 doses over 12 weeks vs elevated natriuretic peptide 13.2 ± 1.4 g/dl); ferritin (283 ± 150 ng/ml
standard of care levels, decreased exercise vs 79 ng/ml); Tsat (27 ± 8% vs
capacity, ferritin <100 μg/l or 20.2%); ∆peak VO2 (–0.16 ± 0.387
100–300 μg/l with Tsat <20% vs 1.19 ± 0.389 ml/min/kg); NYHA
class, patient global assessment
Dhoot et al. Ferric carboxymaltose single Symptomatic with CHF (NYHA 12 weeks Peak VO2 (14.9 ± 3.4 vs 12.9 ± LVEF 250
dose vs placebo class II/III), ferritin <100 μg/l or 3.2 ml/min/kg); NYHA class (class I:
100–300 μg/l with Tsat <20%, 74.3% vs 48.6%, class II: 25.7%
minimum haemoglobin cut-off vs 51.4%); 6MWD (502.1 ± 70 m vs
8 g/dl 455.5 ± 52.3 m); MLHFQ (46.9 ± 15.7
vs 47.9 ± 15.1)
AFFIRM-AHF Ferric carboxymaltose Patients hospitalized for acute HF 52 weeks ∆Haemoglobin (0.8 g/dl vs None 251
2–4 doses over 24 weeks with LVEF <50%, ferritin <100 μg/l 0.3 g/dl); ferritin and Tsat (not
vs placebo or 100–299 μg/l with Tsat <20%, reported numerically); risk of
minimum haemoglobin cut-off recurrent hospitalization for HF
8 g/l or cardiovascular death (RR 0.8)
IRONMAN Isomaltoside 1–9 doses over New or established HF, LVEF 2.7 years Risk of recurrent hospitalization 6MWD 252
20 months vs usual care ≤45% in preceding 24 months, (median for HF or cardiovascular death
ferritin <100 μg/l or Tsat <20% with follow-up) (RR 0.76)
minimum haemoglobin cut-off
9 g/dl
Mollace et al. Ferric carboxymaltose 500 mg HFpEF with ID (ferritin <100 μg/l 8 weeks Group 1 only: ferritin (129.1 ± None 253
at 0 and 4 weeks vs placebo or 100–300 μg/l with Tsat <20%), 13.9 ng/ml vs 62.3 ± 12.4 ng/ml);
three groups according to degree 6MWD (354.4 ± 23.7 m vs
of diastolic function: group 1: E/e′ 294.7 ± 20.7 m)
ratio ≥15, group 2: E/e′ ratio 9–14,
group 3: E/e′ ratio ≤8
HEART-FID Ferric carboxymaltose LVEF ≤40% and either 12 months NA Hierarchical 255
2,316 ± 1,366 mg cumulative hospitalization for HF within the outcome that
dose over 3 years vs placebo previous 12 months or elevated included death,
natriuretic peptide levels, ferritin hospitalizations
<100 μg/l or 100–300 μg/l for HF and
with Tsat <20%, minimum 6MWD
haemoglobin cut-off 9 g/l in
women only
6MWD, 6-minute walking distance; CABG, coronary artery bypass graft; CHF, chronic heart failure; CKD, chronic kidney disease; DHF, decompensated heart failure; EQ-5D, European Quality
of Life – Five Dimensions; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; ID, iron deficiency; KCCQ, Kansas City
Cardiomyopathy Questionnaire; LVEF, left ventricular ejection fraction; MLHFQ, Minnesota Living with Heart Failure Questionnaire; NA, not available; sTFR, soluble transferrin receptor;
Tsat, transferrin saturation; VO2, peak oxygen consumption. aMeta-analysis examined outcomes at 1 year from 64 randomized controlled trials with various follow-up times.

Regarding the long-term fate of iron, MRI studies in patients administration269. In patients with systolic CHF and ID, the MRI T2*
undergoing dialysis showed that the iron concentration in the liver signal indicated that the iron concentration is increased in the heart,
(estimated by magnetic resonance T1 and T2*) was increased by iron liver and spleen but not in the skeletal muscle at 3 months after FCM
sucrose administration but not by FCM or iron isomaltoside administra- infusion166. In another study of patients with CHF and ID, MRI T2* val-
tion, whereas the iron concentration in the spleen (estimated by R2* ues indicated an increase in cardiac iron concentration at 7 days that
relaxometry) was most increased by iron sucrose and iron isomaltoside persisted at 30 days, whereas T1 mapping showed an increase at 7 days

Nature Reviews Cardiology


Review article

that was lost at 30 days after FCM infusion263. The discrepant results of One potential pathway for harm is cumulative myocardial iron
these studies indicate that different MRI parameters reflect different loading given that iron delivered to the myocardium persists there
cellular iron pools, with T1 reflecting the labile cellular iron level that for at least 3 months166. Redosing at shorter intervals could result in
rises early as a result of NTBI uptake, and T2* reflecting the totality of prolonged myocardial exposure to NTBI that, in conditions such as
cellular iron or that which is integrated in cellular ferritin stores or in haemochromatosis and β-thalassaemia, underpins the development
the mitochondria. Notwithstanding these technical considerations, the of iron overload cardiomyopathy273,274. The interaction between intra-
short-term and long-term fates of various intravenous iron formula- venous iron and phosphate homeostasis is another important safety
tions in non-erythroid tissues, where iron turnover is far slower than in consideration in the context of repeated dosing because of the risk
the erythroid tissue, warrant further investigation. This research will aid of sustained hypophosphataemia, subsequent hypophosphataemic
our understanding of the basis and time course of any observed ben- osteomalacia and its sequelae, including fractures275. Long-term moni-
efits and identify dosing intervals that safeguard against cumulative toring of NTBI levels, phosphate levels and myocardial iron concentra-
iron loading into non-erythroid tissues. tion should form part of future trials of intravenous iron therapy that
use repeated dosing regimens.
Inflammation and the response to intravenous iron. One of the reasons
for using intravenous rather than oral iron formulations in patients with When intravenous iron should be avoided. Some evidence suggests
HF is to bypass the inhibitory effect of hepcidin on absorption in the that intravenous iron therapy should not be used in the setting of
gut. However, hepcidin also controls iron efflux from macrophages, myocardial reperfusion (after myocardial infarction, percutaneous
which are responsible for the breakdown and release of most of the coronary intervention or CABG surgery). In this setting, preclinical
iron from the carbohydrate shell. Consequently, elevation of serum models provide proof of concept that iron therapy promotes myo-
hepcidin levels would result in iron eventually becoming trapped in cardial ischaemia–reperfusion injury276,277. Moreover, in patients with
macrophages (Fig. 5). This trapping could be compounded by the fact ST-segment elevation myocardial infarction, ID was found to be cou-
that the intravenous iron itself induces a rapid and marked increase in pled with smaller areas of myocardial ischaemia–reperfusion injury
hepcidin level36,57. A study in which the homing of supplemented iron and better in-hospital outcomes, whereas NTBI levels correlated with
was compared between rat models of ID anaemia and ACD supports this markers of cardiac injury, particularly in patients with evidence of
idea270. In the absence of treatment, the liver and spleen of rats with ID microvascular obstruction and haemorrhage278,279. In CABG surgery,
anaemia were iron depleted, whereas those of rats with ACD showed iron iron chelation with desferroxamine was found to ameliorate lipid per-
retention in Kupffer cells in the liver and splenic macrophages. In rats oxidation and protect the myocardium against ischaemia–reperfusion
with ID anaemia, intravenous iron therapy reversed anaemia and resulted injury280. Accordingly, clinical and preclinical data support the notion
in the appearance of iron in Kupffer cells and hepatocytes. By contrast, that pre-emptive or delayed iron supplementation might be safer than
intravenous iron therapy did not correct anaemia in rats with ACD and acute supplementation281–287.
resulted in iron retention in Kupffer cells but not in hepatocytes270. Another setting in which intravenous iron therapy might be det-
One implication of these findings is that, in some patients, the rise rimental is arrhythmias, including atrial fibrillation. Polymorphisms
in ferritin levels after intravenous iron infusion reflects the extent of associated with raised serum levels of iron, Tsat and ferritin are also asso-
iron trapping in macrophages (which are the primary source of serum ciated with an increased risk of atrial fibrillation288. Furthermore,
ferritin) rather than repletion of liver iron stores. Another concern raised serum ferritin concentrations are associated with atrial fibril-
relates to the trapping of iron in lung epithelial cells and macrophages, lation risk in the general population73. In patients with haemophilia A,
which has the potential to exacerbate lung fibrosis in patients with serial phlebotomy resulted in remission from symptomatic atrial
both HF and COPD. Comparing macrophage iron trapping after intra- fibrillation289. Furthermore, arrythmias affect ~16% of patients with
venous iron therapy between patients with absolute ID and those with haemochromatosis290,291. The mechanistic links between increased iron
functional ID will help to determine the extent to which macrophage levels and arrythmias seem to relate to abnormal calcium handling,
iron trapping relates to the recovery of haematological parameters specifically the effects of iron on L-type calcium channels292–294. The
and the improvement in functional outcomes. ongoing IRON-AF trial295 of intravenous iron therapy in patients with
atrial fibrillation and ID should provide insights as to whether iron
Safety of repeated intravenous iron dosing. Repeated dosing supplementation is beneficial or harmful in this setting. Finally, in
with intravenous iron is increasingly being used to maintain ferritin patients with HF and COPD, the potential for intravenous iron therapy
and/or haemoglobin levels in the normal range. In iron therapy trials to compound lung iron loading and fibrosis should be considered.
of patients with HF, the maximum cumulative dose of intravenous iron
resulting from repeated infusion ranges from 2 g to 13 g, equivalent Cost-effectiveness of intravenous iron. In the setting of CHF, analyses
to between one and four times the total amount of iron in the body of in which the reduced risk of hospitalization and recurrent HF have been
healthy individuals245,247,251–253. One pertinent question is why such a considered suggest that intravenous iron therapy is cost-effective241,284.
large dose of iron is needed to maintain haemoglobin or ferritin levels in However, no cost-effectiveness analyses have been performed on the
the normal range in the absence of substantial blood loss. One possibil- comparison between intravenous and oral iron therapy or factor-
ity is that patients requiring repeated dosing are those in whom iron is ing in the financial cost and potential harm of repeated intravenous
trapped in macrophages, potentially owing to a high hepcidin level, as iron dosing.
discussed previously270. Evidence from patients receiving dialysis sug-
gests that intravenous iron dosing regimens of ≥300 mg per month are Opportunities in the management of ID in HF
associated with increased mortality271,272. In patients with HF, repeated Our understanding of iron homeostasis has grown rapidly since the
iron dosing seems to be well tolerated in the timescale of the trials turn of the century, providing a mechanistic basis for the behaviour of
(typically 6–20 months) but its long-term safety remains unexplored. various iron markers, the physiological role of iron outside the erythron

Nature Reviews Cardiology


Review article

Fig. 5 | The effects of inflammation


a No inflammation NTBI
on the fate of intravenous iron
Fe
supplementation. Comparison of
iron handling after intravenous iron
infusion between non-inflammatory
Spleen Liver Lung and inflammatory states. a, In non-
Alveolar
Splenic Kupffer inflammatory states, when hepcidin
Hepatocyte macrophage
macrophage cell is not elevated, iron delivered to
Lung
Fe Fe epithelium macrophages is readily mobilized
Fe ↑Fe
Fe into the circulation to meet demands
of the erythroid compartment as well
as those of the skeletal muscle and
myocardium, and any excess iron
Mobilization Mobilization Mobilization Storage Mobilization can become stored in hepatocytes,
where it is also readily mobilizable
when needed. b, In inflammatory
↑↑ Tf(Fe3+)
states, elevated hepcidin levels
prevent the mobilization of iron from
macrophages, causing iron trapping.
Ferroportin Iron can also become trapped in
↑Fe ↑Fe alveolar macrophages. At the same
Fe Transferrin
time, any iron stored in hepatocytes is
↑↑ Haemoglobin Myotubule Cardiomyocyte not mobilizable. Overall, iron trapping
TFR
in inflammatory states reduces the
Erythroblast Intravenous
Bone marrow Skeletal muscle Myocardium uptake of iron into the erythroid
iron drug
compartment, myocardial and skeletal
NTBI transporter
muscles, resulting in a poor response
Flux reduced in
to intravenous iron therapy. NTBI,
inflammation
b Inflammation NTBI non-transferrin-bound iron; Tf(Fe3+),
Fe transferrin-bound iron; TFR, transferrin
receptor.

Spleen Liver Lung


Alveolar
Splenic Kupffer Hepatocyte macrophage
macrophage cell
Lung
↑Fe ↑Fe epithelium
↑Fe
↑Fe

Hepcidin

No
No mobilization No mobilization mobilization No storage No mobilization

↑ Tf(Fe3+)

↑Fe ↑Fe

↑ Haemoglobin Myotubule Cardiomyocyte

Erythroblast
Bone marrow Skeletal muscle Myocardium

and the handling of exogenous iron in the body. This growing body of in particular has proved to be a problematic iron marker due to its
knowledge presents various untapped opportunities to improve the inability to resolve true repletion of iron stores from macrophage iron
management of ID in patients with HF. trapping255–257. Existing ferritin thresholds should be re-evaluated; for
instance, the ferritin values below which absorption increases can be
Improve interpretation of existing iron markers determined with isotopic iron tracing. Using this approach, Galetti et al.
In HF, the current ESC definition of ID does not predict worse prog- found that a ferritin cut-off of 50 μg/l denoted “physiological iron defi-
nosis or a favourable response to iron supplementation137. Ferritin ciency” in young women296. A similar strategy could be used to refine

Nature Reviews Cardiology


Review article

ferritin thresholds for individuals with varying degrees of inflammation. used in conjunction with oral or intravenous iron supplementation
Analysis of the ferritin light-to-heavy-chain ratio and its glycosylation to meet the concomitant increase in erythroid iron demand, and this
status could also improve the resolving power of ferritin62,63. Changes combinatorial approach has been shown to be superior to iron therapy
in Tsat should be interpreted in the context of changes in both serum alone in patients with HF and anaemia302,303. However, recombinant
iron and serum transferrin levels. The presence of liver injury, which erythropoietin has been linked with an increased risk of recurrent HF
can cause both ferritin leakage and impairment of transferrin biosyn- and death304. Therefore, one alternative is to stimulate endogenous
thesis, should also be considered64,79,80. Finally, as a shared regulator erythropoietin production with HIF prolyl hydroxylase inhibitors
of ferritin, transferrin and erythropoietin, TNF could prove useful in developed to treat anaemia. Indeed, molidustat has already been shown
distinguishing ACD from ID anaemia66,68,83,84. to suppress hepcidin in patients with anaemia associated with CKD305.
Recombinant erythroferrone itself could be developed to normalize
Develop biomarkers of unmet iron need hepcidin levels in functional ID, with the advantage that erythroferrone,
sTFR is emerging as a promising marker of the unmet need for iron by unlike erythropoietin, does not place additional erythroid demand on
the erythron because it is derived from the shedding of TFR1 from the iron resources and does not adversely affect HF outcomes. In preclini-
surface of erythroblasts, where TFR1 expression is directly regulated cal models, recombinant erythroferrone inhibits hepcidin, prevents its
by IRP1 according to intracellular iron availability95,103,297. There are no induction by BMP6 and IL-6, and contributes to recovery from ACD29,30.
established biomarkers of the unmet need for iron outside the erythron. Finally, hepcidin could be targeted directly. One hepcidin antagonist
The regulation of endothelin 1 by the iron status of pulmonary arterial (PRS-080#22) has been tested in phase I trial in healthy volunteers and
smooth muscle cells suggests that endothelin 1 should be explored in patients with CKD undergoing dialysis, with promising effects on iron
further as a candidate ID marker for the pulmonary circulation38,213. parameters306. In a primate model, this antagonist achieved sustained
Metabolomic and proteomic studies are warranted to identify circu- hepcidin suppression and elevation of plasma iron markers307.
lating biomarkers of the unmet need for iron in the myocardium and
skeletal muscle. In the meantime, iron quantification techniques based Conclusions
on cardiac magnetic resonance T1, T2 and T2* (traditionally used for the Having an unmet need for iron is detrimental in a broad range of cardio-
detection of iron overload) should be optimized to detect myocardial vascular conditions, particularly in HF. Although the unmet need for
iron depletion. This approach could be used to identify patients with a iron by the erythron is readily diagnosed through measurement of hae-
truly unmet need for iron in the myocardium, guide dosing regimens moglobin and sTFR concentrations, diagnosis of the unmet needs for
and safeguard against excess iron loading. iron by non-erythroid tissues remains extremely challenging. Current
circulating iron markers, although useful in the general population, are
Re-evaluate oral and intravenous iron therapies much harder to interpret in patients with HF, because of the confound-
The benefits of oral iron therapies should be re-evaluated in the ing effects of inflammation and hypoxia, hepatic, renal and respiratory
light of updated guidelines on alternate-day dosing and the advent comorbidities, and the use of certain medications. The shortcomings
of new slow-release formulations, such as sucrosomial iron, which of these markers in identifying those with a truly unmet need for iron
seem to be less sensitive than earlier formulations to the effects have probably limited the success of oral and intravenous iron thera-
of inflammation218,219. Intravenous iron therapies should also be pies in patients with HF and other cardiovascular conditions. Dissect-
re-evaluated in the context of evidence for differential iron handling ing, at a more fundamental level, the mechanisms and importance of
in patients with absolute ID and those with functional ID as well as the tissue-level iron control can help to identify biomarkers of an unmet
potential for macrophage iron trapping in the latter setting. need for iron outside the erythron. These biomarkers would transform
the management of iron status, from a one-size-fits-all approach to
Develop alternatives to iron supplementation personalized strategies, tailored quantitatively and qualitatively to the
An alternative approach to supplementing iron is to target the homeo- specific needs of each patient.
static pathways that control iron levels and distribution in the body. Until novel biomarkers are identified, existing iron therapies
Normalizing hepcidin in functional ID would restore the ability of the should be re-evaluated in light of new data on the physiological han-
gut to absorb iron, unlock the iron that is trapped in macrophages and dling of iron supplements, including the effects of the type of formu-
hepatocytes, and allow tissues that express ferroportin (such as the lation and the speed of iron release on hepcidin, and the modifying
myocardium and pulmonary vasculature) to maintain normal homeo- effects of anaemia and inflammation. This re-evaluation should also
static control of the LIP35–38. Hepcidin levels can be normalized with the consider the possibility that pathways of benefit differ between patients
use of some anti-inflammatory drugs; for example, those against IL-1 or according to the specific nature of their unmet need for iron. Equally
IL-6. The IL-1β inhibitor canakinumab has been shown to both reverse important is the identification of predictors of poor response to iron
and prevent anaemia in patients with previous myocardial infarction and supplementation to minimize repeated dosing of poor responders, for
elevated CRP level. These effects were associated with a dose-dependent which the availability of long-term safety data is limited. In functional
reduction in hospitalization for HF and HF-related mortality298,299. Hep- ID, greater attention should be given to approaches that can un-trap
cidin was one of the most downregulated proteins in the active group iron such as targeting the inflammatory pathways that induce hepcidin
of a trial involving the IL-6 receptor antagonist tocilizumab in patients or leveraging the ability of the erythroferrone axis to overcome the
with non-ST-segment elevation myocardial infarction143. effect of inflammation on hepcidin. Cardiovascular medicine has led
As an alternative to anti-inflammatory drugs, hepcidin could be the way both in the recognition of NAID as an important clinical target
normalized by leveraging the dominant inhibitory effects of erythro- and in embracing novel iron therapies and is therefore well placed to
poietin and erythroferrone. In healthy volunteers, microdosing with tackle these future challenges and realize the untapped opportunities.
recombinant human erythropoietin is sufficient to increase erythro-
ferrone levels and suppress hepcidin300,301. Erythropoietin could be Published online: xx xx xxxx

Nature Reviews Cardiology


Review article

References 34. Das, S. K. & Oudit, G. Y. Voltage-gated Ca2+ channels as key mediators of iron-transport
1. O’Meara, E. et al. Clinical correlates and consequences of anemia in a broad spectrum and iron-overload cardiomyopathy: L-type vs. T-type Ca+ channels. Eur. J. Haematol. 88,
of patients with heart failure: results of the Candesartan in Heart Failure: Assessment of 476–477 (2012).
Reduction in Mortality and Morbidity (CHARM) program. Circulation 113, 986–994 35. Lakhal-Littleton, S. et al. Cardiac ferroportin regulates cellular iron homeostasis and is
(2006). important for cardiac function. Proc. Natl Acad. Sci. USA 112, 3164–3169 (2015).
2. Graham, F. J., Friday, J. M., Pellicori, P., Greenlaw, N. & Cleland, J. G. Assessment of 36. Lakhal-Littleton, S. et al. An essential cell-autonomous role for hepcidin in cardiac iron
haemoglobin and serum markers of iron deficiency in people with cardiovascular homeostasis. eLife 5, e19804 (2016).
disease. Heart 109, 1294–1301 (2023). 37. Loick, P. et al. Protective role for smooth muscle cell hepcidin in abdominal aortic
3. Lindberg, F. et al. Iron deficiency in heart failure: screening, prevalence, incidence and aneurysm. Arterioscler. Thromb. Vasc. Biol. 43, 713–725 (2023).
outcome data from the Swedish Heart Failure Registry and the Stockholm CREAtinine 38. Lakhal-Littleton, S. et al. Intracellular iron deficiency in pulmonary arterial smooth
Measurements collaborative project. Eur. J. Heart Fail. 25, 1270–1280 (2023). muscle cells induces pulmonary arterial hypertension in mice. Proc. Natl Acad. Sci. USA
4. Ilbert, M. & Bonnefoy, V. Insight into the evolution of the iron oxidation pathways. 116, 13122–13130 (2019).
Biochim. Biophys. Acta 1827, 161–175 (2013). 39. Noetzli, L. J., Papudesi, J., Coates, T. D. & Wood, J. C. Pancreatic iron loading predicts
5. Pierre, J. L. & Fontecave, M. Iron and activated oxygen species in biology: the basic cardiac iron loading in thalassemia major. Blood 114, 4021–4026 (2009).
chemistry. Biometals 12, 195–199 (1999). 40. Steinbicker, A. U. & Muckenthaler, M. U. Out of balance-systemic iron homeostasis in
6. FAO, IFAD, UNICEF, WFP, WHO. The State of Food Security and Nutrition in the World 2017. iron-related disorders. Nutrients 5, 3034–3061 (2013).
Building Resilience for Peace and Food Security 1–109 (FAO, 2017). 41. Loreal, O. et al. Iron as a therapeutic target in HFE-related hemochromatosis: usual and
7. von Haehling, S. et al. Prevalence and clinical impact of iron deficiency and anaemia novel aspects. Pharmaceuticals 11, 131 (2018).
among outpatients with chronic heart failure: the PrEP Registry. Clin. Res. Cardiol. 106, 42. Huebers, H. A. & Finch, C. A. The physiology of transferrin and transferrin receptors.
436–443 (2017). Physiol. Rev. 67, 520–582 (1987).
8. Klip, I. T. et al. Iron deficiency in chronic heart failure: an international pooled analysis. 43. Cohen, L. A. et al. Serum ferritin is derived primarily from macrophages through a
Am. Heart J. 165, 575–582.e3 (2013). nonclassical secretory pathway. Blood 116, 1574–1584 (2010).
9. Nunez, J. et al. Iron deficiency and risk of early readmission following a hospitalization for 44. Ferring-Appel, D., Hentze, M. W. & Galy, B. Cell-autonomous and systemic context-
acute heart failure. Eur. J. Heart Fail. 18, 798–802 (2016). dependent functions of iron regulatory protein 2 in mammalian iron metabolism.
10. Martens, P. et al. Impact of iron deficiency on exercise capacity and outcome in heart Blood 113, 679–687 (2009).
failure with reduced, mid-range and preserved ejection fraction. Acta Cardiol. 73, 115–123 45. Watanabe, K. et al. Iron content of rat serum ferritin. J. Vet. Med. Sci. 63, 587–589 (2001).
(2018). 46. Nemeth, E. & Ganz, T. Hepcidin-ferroportin interaction controls systemic iron
11. Jankowska, E. A. et al. Iron deficiency: an ominous sign in patients with systolic chronic homeostasis. Int. J. Mol. Sci. 22, 6493 (2021).
heart failure. Eur. Heart J. 31, 1872–1880 (2010). 47. Ganz, T. Cellular iron: ferroportin is the only way out. Cell Metab. 1, 155–157 (2005).
12. Palau, P. et al. Iron deficiency and short-term adverse events in patients with 48. Mohammad, G., Matakidou, A., Robbins, P. A. & Lakhal-Littleton, S. The kidney hepcidin/
decompensated heart failure. Clin. Res. Cardiol. 110, 1292–1298 (2021). ferroportin axis controls iron reabsorption and determines the magnitude of kidney and
13. Weidmann, H. et al. Iron metabolism contributes to prognosis in coronary artery disease: systemic iron overload. Kidney Int. 100, 559–569 (2021).
prognostic value of the soluble transferrin receptor within the atherogene study. J. Am. 49. Wareing, M., Ferguson, C. J., Green, R., Riccardi, D. & Smith, C. P. In vivo characterization
Heart Assoc. 9, e015480 (2020). of renal iron transport in the anaesthetized rat. J. Physiol. 524, 581–586 (2000).
14. Chen, W., Lin, G., Dai, C. & Xu, K. Predictive value of serum iron on heart failure in patients 50. Napolitano, M. et al. Iron-dependent erythropoiesis in women with excessive menstrual
with acute ST-segment elevation myocardial infarction. Clin. Cardiol. 46, 449–453 blood losses and women with normal menses. Ann. Hematol. 93, 557–563 (2014).
(2023). 51. Hytten, F. E., Cheyne, G. A. & Klopper, A. I. Iron loss at menstruation. J. Obstet. Gynaecol.
15. Lawen, A. & Lane, D. J. Mammalian iron homeostasis in health and disease: uptake, Br. Commonw. 71, 255–259 (1964).
storage, transport, and molecular mechanisms of action. Antioxid. Redox Signal. 18, 52. Sangkhae, V., Fisher, A. L., Ganz, T. & Nemeth, E. Iron homeostasis during pregnancy:
2473–2507 (2013). maternal, placental, and fetal regulatory mechanisms. Annu. Rev. Nutr. 43, 279–300
16. Oski, F. A. Iron deficiency in infancy and childhood. N. Engl. J. Med. 329, 190–193 (1993). (2023).
17. Siddappa, A. M., Rao, R., Long, J. D., Widness, J. A. & Georgieff, M. K. The assessment 53. Lakhal-Littleton, S. Advances in understanding the crosstalk between mother and fetus
of newborn iron stores at birth: a review of the literature and standards for ferritin on iron utilization. Semin. Hematol. 58, 153–160 (2021).
concentrations. Neonatology 92, 73–82 (2007). 54. Moretti, D. et al. Oral iron supplements increase hepcidin and decrease iron absorption
18. Kohgo, Y., Ikuta, K., Ohtake, T., Torimoto, Y. & Kato, J. Body iron metabolism and from daily or twice-daily doses in iron-depleted young women. Blood 126, 1981–1989
pathophysiology of iron overload. Int. J. Hematol. 88, 7–15 (2008). (2015).
19. Cabantchik, Z. I. Labile iron in cells and body fluids: physiology, pathology, 55. Giordano, G., Napolitano, M., Di Battista, V. & Lucchesi, A. Oral high-dose sucrosomial
and pharmacology. Front. Pharmacol. 5, 45 (2014). iron vs intravenous iron in sideropenic anemia patients intolerant/refractory to iron
20. Knutson, M. D. Non-transferrin-bound iron transporters. Free Radic. Biol. Med. 133, sulfate: a multicentric randomized study. Ann. Hematol. 100, 2173–2179 (2021).
101–111 (2019). 56. Remacha, A. et al. Guidelines on haemovigilance of post-transfusional iron overload.
21. Frazer, D. M. et al. A rapid decrease in the expression of DMT1 and Dcytb but not Ireg1 Blood Transfus. 11, 128–139 (2013).
or hephaestin explains the mucosal block phenomenon of iron absorption. Gut 52, 57. Garbowski, M. W. et al. Intravenous iron preparations transiently generate
340–346 (2003). non-transferrin-bound iron from two proposed pathways. Haematologica 106,
22. Philpott, C. C. Coming into view: eukaryotic iron chaperones and intracellular iron 2885–2896 (2021).
delivery. J. Biol. Chem. 287, 13518–13523 (2012). 58. Geisser, P. & Burckhardt, S. The pharmacokinetics and pharmacodynamics of iron
23. Rouault, T. & Klausner, R. Regulation of iron metabolism in eukaryotes. Curr. Top. Cell Regul. preparations. Pharmaceutics 3, 12–33 (2011).
35, 1–19 (1997). 59. Murray, M. J., Murray, A. B., Murray, M. B. & Murray, C. J. The adverse effect of iron
24. Arosio, P., Carmona, F., Gozzelino, R., Maccarinelli, F. & Poli, M. The importance of repletion on the course of certain infections. BMJ 2, 1113–1115 (1978).
eukaryotic ferritins in iron handling and cytoprotection. Biochem. J. 472, 1–15 (2015). 60. Bullen, J. J., Spalding, P. B., Ward, C. G. & Gutteridge, J. M. Hemochromatosis, iron and
25. Shi, R., Hou, W., Wang, Z. Q. & Xu, X. Biogenesis of iron-sulfur clusters and their role in septicemia caused by Vibrio vulnificus. Arch. Intern. Med. 151, 1606–1609 (1991).
DNA metabolism. Front. Cell Dev. Biol. 9, 735678 (2021). 61. Lakhal-Littleton, S. & Robbins, P. A. The interplay between iron and oxygen homeostasis
26. McDonald, R., Hegenauer, J., Sucec, A. & Saltman, P. Effects of iron deficiency and with a particular focus on the heart. J. Appl. Physiol. 123, 967–973 (2017).
exercise on myoglobin in rats. Eur. J. Appl. Physiol. Occup. Physiol. 52, 414–419 (1984). 62. Cragg, S. J., Wagstaff, M. & Worwood, M. Detection of a glycosylated subunit in human
27. Muckenthaler, M. U., Rivella, S., Hentze, M. W. & Galy, B. A red carpet for iron metabolism. serum ferritin. Biochem. J. 199, 565–571 (1981).
Cell 168, 344–361 (2017). 63. Lambotte, O. et al. High ferritin and low glycosylated ferritin may also be a marker of
28. Kohgo, Y. et al. Serum transferrin receptor as a new index of erythropoiesis. Blood 70, excessive macrophage activation. J. Rheumatol. 30, 1027–1028 (2003).
1955–1958 (1987). 64. Moris, W., Verhaegh, P., Jonkers, D., Deursen, C. V. & Koek, G. Hyperferritinemia in
29. Arezes, J. et al. Erythroferrone inhibits the induction of hepcidin by BMP6. Blood 132, nonalcoholic fatty liver disease: iron accumulation or inflammation? Semin. Liver Dis. 39,
1473–1477 (2018). 476–482 (2019).
30. Kautz, L., Jung, G., Nemeth, E. & Ganz, T. Erythroferrone contributes to recovery from 65. Yenson, P. R., Yoshida, E. M., Li, C. H., Chung, H. V. & Tsang, P. W. Hyperferritinemia in
anemia of inflammation. Blood 124, 2569–2574 (2014). the Chinese and Asian community: a retrospective review of the University of British
31. Bothwell, T. H. et al. In: Iron Metabolism in Man: Clinical Estimation of Body Iron Columbia experience. Can. J. Gastroenterol. 22, 37–40 (2008).
Stores pp 88–93 (eds Bothwell, T. H., Charlton, R., Cook, J. D. & Finch, C. A.) 327–349 66. Konijn, A. M., Carmel, N., Levy, R. & Hershko, C. Ferritin synthesis in inflammation.
(Blackwell Scientific Publications, 1979). II. Mechanism increased ferritin synthesis. Br. J. Haematol. 49, 361–370 (1981).
32. de Swart, L. et al. Second international round robin for the quantification of serum 67. Pietsch, E. C., Chan, J. Y., Torti, F. M. & Torti, S. V. Nrf2 mediates the induction of ferritin H
non-transferrin-bound iron and labile plasma iron in patients with iron-overload in response to xenobiotics and cancer chemopreventive dithiolethiones. J. Biol. Chem.
disorders. Haematologica 101, 38–45 (2016). 278, 2361–2369 (2003).
33. Fanni, D. et al. Effects of iron and copper overload on the human liver: an ultrastructural 68. Kernan, K. F. & Carcillo, J. A. Hyperferritinemia and inflammation. Int. Immunol. 29,
study. Curr. Med. Chem. 21, 3768–3774 (2014). 401–409 (2017).

Nature Reviews Cardiology


Review article

69. Karaboyas, A. et al. Association between serum ferritin and mortality: findings from the 102. Schmidt, P. J., Toran, P. T., Giannetti, A. M., Bjorkman, P. J. & Andrews, N. C. The transferrin
USA, Japan and European Dialysis Outcomes and Practice Patterns Study. Nephrol. Dial. receptor modulates Hfe-dependent regulation of hepcidin expression. Cell Metab. 7,
Transpl. 33, 2234–2244 (2018). 205–214 (2008).
70. Kalantar-Zadeh, K., Don, B. R., Rodriguez, R. A. & Humphreys, M. H. Serum ferritin is 103. Infusino, I., Braga, F., Dolci, A. & Panteghini, M. Soluble transferrin receptor (sTfR)
a marker of morbidity and mortality in hemodialysis patients. Am. J. Kidney Dis. 37, and sTfR/log ferritin index for the diagnosis of iron-deficiency anemia. A meta-analysis.
564–572 (2001). Am. J. Clin. Pathol. 138, 642–649 (2012).
71. Park, K. S. et al. Serum ferritin predicts mortality regardless of inflammatory and 104. Tussing-Humphreys, L. M. et al. Elevated systemic hepcidin and iron depletion in obese
nutritional status in patients starting dialysis: a prospective cohort study. Blood Purif. premenopausal females. Obesity 18, 1449–14569 (2010).
40, 209–217 (2015). 105. Verga Falzacappa, M. V. et al. STAT3 mediates hepatic hepcidin expression and its
72. Ellervik, C., Marott, J. L., Tybjaerg-Hansen, A., Schnohr, P. & Nordestgaard, B. G. Total and inflammatory stimulation. Blood 109, 353–358 (2007).
cause-specific mortality by moderately and markedly increased ferritin concentrations: 106. Oliveira, S. J. et al. ER stress-inducible factor CHOP affects the expression of hepcidin
general population study and metaanalysis. Clin. Chem. 60, 1419–1428 (2014). by modulating C/EBPalpha activity. PLoS One 4, e6618 (2009).
73. Mikkelsen, L. F., Nordestgaard, B. G., Schnohr, P. & Ellervik, C. Increased ferritin 107. Krause, A. et al. LEAP-1, a novel highly disulfide-bonded human peptide, exhibits
concentration and risk of atrial fibrillation and heart failure in men and women: three antimicrobial activity. FEBS Lett. 480, 147–150 (2000).
studies of the Danish general population including 35799 individuals. Clin. Chem. 65, 108. Kautz, L. et al. Identification of erythroferrone as an erythroid regulator of iron
180–188 (2019). metabolism. Nat. Genet. 46, 678–684 (2014).
74. Holay, M. P., Choudhary, A. A. & Suryawanshi, S. D. Serum ferritin-a novel risk factor in 109. Kautz, L. et al. Erythroferrone contributes to hepcidin suppression and iron overload in a
acute myocardial infarction. Indian Heart J. 64, 173–177 (2012). mouse model of β-thalassemia. Blood 126, 2031–2037 (2015).
75. Brissot, P., Bernard, D. G., Brissot, E., Loreal, O. & Troadec, M. B. Rare anemias due to 110. Zaman, B. A., Rasool, S. O. & Abdo, J. M. The effect of erythroferrone suppression by
genetic iron metabolism defects. Mutat. Res. Rev. Mutat. Res. 777, 52–63 (2018). transfusion on the erythropoietin-erythroferrone-hepcidin axis in transfusion-dependent
76. Yang, Y. et al. Changes of iron metabolism during pregnancy and the establishment thalassaemia: a pre-post cohort study. Br. J. Haematol. 201, 547–551 (2023).
of reference intervals for pregnant Chinese women. Ann. Clin. Biochem. 56, 556–563 111. Stoffel, N. U. et al. The opposing effects of acute inflammation and iron deficiency
(2019). anemia on serum hepcidin and iron absorption in young women. Haematologica 104,
77. Rolfs, A., Kvietikova, I., Gassmann, M. & Wenger, R. H. Oxygen-regulated transferrin 1143–1149 (2019).
expression is mediated by hypoxia-inducible factor-1. J. Biol. Chem. 272, 20055–20062 112. Silvestri, L., Pagani, A. & Camaschella, C. Furin-mediated release of soluble hemojuvelin:
(1997). a new link between hypoxia and iron homeostasis. Blood 111, 924–931 (2008).
78. Baynes, R. D., Meyer, T. E., Bothwell, T. H. & McNamara, L. A screening test for detecting 113. Lakhal, S. et al. Regulation of type II transmembrane serine proteinase TMPRSS6 by
iron overload in population studies. S. Afr. Med. J. 74, 167–169 (1988). hypoxia-inducible factors: new link between hypoxia signaling and iron homeostasis.
79. Viveiros, A. et al. Transferrin as a predictor of survival in cirrhosis. Liver Transpl. 24, J. Biol. Chem. 286, 4090–4097 (2011).
343–351 (2018). 114. Liu, W. et al. Lactate modulates iron metabolism by binding soluble adenylyl cyclase.
80. Bergmann, O. M. et al. Altered expression of iron regulatory genes in cirrhotic human Cell Metab. 35, 1597–1612.e6 (2023).
livers: clues to the cause of hemosiderosis? Lab. Invest. 88, 1349–1357 (2008). 115. Troutt, J. S., Butterfield, A. M. & Konrad, R. J. Hepcidin-25 concentrations are
81. Hancock, D. E., Onstad, J. W. & Wolf, P. L. Transferrin loss into the urine with hypochromic, markedly increased in patients with chronic kidney disease and are inversely
microcytic anemia. Am. J. Clin. Pathol. 65, 73–78 (1976). correlated with estimated glomerular filtration rates. J. Clin. Lab. Anal. 27, 504–510
82. Howard, R. L., Buddington, B. & Alfrey, A. C. Urinary albumin, transferrin and iron (2013).
excretion in diabetic patients. Kidney Int. 40, 923–926 (1991). 116. Washida, N. et al. Impact of switching from darbepoetin alfa to epoetin beta pegol on
83. Jain, S., Gautam, V. & Naseem, S. Acute-phase proteins: as diagnostic tool. J. Pharm. iron utilization and blood pressure in peritoneal dialysis patients. Ther. Apher. Dial. 19,
Bioallied Sci. 3, 118–127 (2011). 450–456 (2015).
84. Fahmy, M. & Young, S. P. Modulation of iron metabolism in monocyte cell line U937 by 117. Paliege, A. et al. Hypoxia-inducible factor-2α-expressing interstitial fibroblasts are the
inflammatory cytokines: changes in transferrin uptake, iron handling and ferritin mRNA. only renal cells that express erythropoietin under hypoxia-inducible factor stabilization.
Biochem. J. 296, 175–181 (1993). Kidney Int. 77, 312–318 (2010).
85. Gabay, C. & Kushner, I. Acute-phase proteins and other systemic responses to 118. Rezvani, A. et al. Serum levels of erythropoietin in patients with chronic obstructive
inflammation. N. Engl. J. Med. 340, 448–454 (1999). pulmonary disease and anemia. Sci. Rep. 13, 6990 (2023).
86. Griffiths, J. D. et al. Acute changes in iron metabolism following myocardial infarction. 119. Ren, X., Dorrington, K. L., Maxwell, P. H. & Robbins, P. A. Effects of desferrioxamine on
Am. J. Clin. Pathol. 84, 649–654 (1985). serum erythropoietin and ventilatory sensitivity to hypoxia in humans. J. Appl. Physiol.
87. Van der Schouw, Y. T. et al. Iron status in the acute phase and six weeks after myocardial 89, 680–686 (2000).
infarction. Free Radic. Biol. Med. 8, 47–53 (1990). 120. Wilkinson, N. & Pantopoulos, K. IRP1 regulates erythropoiesis and systemic iron
88. Ridefelt, P., Larsson, A., Rehman, J. U. & Axelsson, J. Influences of sleep and the circadian homeostasis by controlling HIF2α mRNA translation. Blood 122, 1658–1668 (2013).
rhythm on iron-status indices. Clin. Biochem. 43, 1323–1328 (2010). 121. Ghosh, M. C. et al. Deletion of iron regulatory protein 1 causes polycythemia and
89. Smith, K. D. Iron metabolism at the host pathogen interface: lipocalin 2 and the pulmonary hypertension in mice through translational derepression of HIF2α.
pathogen-associated iroA gene cluster. Int. J. Biochem. Cell Biol. 39, 1776–1780 (2007). Cell Metab. 17, 271–281 (2013).
90. Nemeth, E. et al. Hepcidin, a putative mediator of anemia of inflammation, is a type II 122. Pang, Y. et al. A novel splicing site IRP1 somatic mutation in a patient with
acute-phase protein. Blood 101, 2461–2463 (2003). pheochromocytoma and JAK2V617F positive polycythemia vera: a case report.
91. Sangkhae, V. & Nemeth, E. Regulation of the iron homeostatic hormone hepcidin. BMC Cancer 18, 286 (2018).
Adv. Nutr. 8, 126–136 (2017). 123. Paulson, R. F., Ruan, B., Hao, S. & Chen, Y. Stress erythropoiesis is a key inflammatory
92. Schwartz, A. J. et al. Hepatic hepcidin/intestinal HIF-2α axis maintains iron absorption response. Cells 9, 634 (2020).
during iron deficiency and overload. J. Clin. Invest. 129, 336–348 (2019). 124. Kim, A. et al. A mouse model of anemia of inflammation: complex pathogenesis with
93. Goetze, O. et al. Adaptation of iron transport and metabolism to acute high-altitude partial dependence on hepcidin. Blood 123, 1129–1136 (2014).
hypoxia in mountaineers. Hepatology 58, 2153–2162 (2013). 125. Hao, S. et al. Gdf15 regulates murine stress erythroid progenitor proliferation and the
94. Pfeiffer, C. M. & Looker, A. C. Laboratory methodologies for indicators of iron status: development of the stress erythropoiesis niche. Blood Adv. 3, 2205–2217 (2019).
strengths, limitations, and analytical challenges. Am. J. Clin. Nutr. 106, 1606S–1614S 126. World Health Organization. WHO Guideline on Use of Ferritin Concentrations to Assess
(2017). Iron Status in Individuals and Populations WHO Guidelines Approved by the Guidelines
95. Chitambar, C. R., Loebel, A. L. & Noble, N. A. Shedding of transferrin receptor from rat Review Committee (WHO, 2020).
reticulocytes during maturation in vitro: soluble transferrin receptor is derived from 127. Pavord, S. et al. UK guidelines on the management of iron deficiency in pregnancy.
receptor shed in vesicles. Blood 78, 2444–2450 (1991). Br. J. Haematol. 188, 819–830 (2020).
96. Pan, B. T. & Johnstone, R. M. Fate of the transferrin receptor during maturation of sheep 128. Batchelor, E. K., Kapitsinou, P., Pergola, P. E., Kovesdy, C. P. & Jalal, D. I. Iron deficiency
reticulocytes in vitro: selective externalization of the receptor. Cell 33, 967–978 (1983). in chronic kidney disease: updates on pathophysiology, diagnosis, and treatment.
97. Daschner, M., Mehls, O. & Schaefer, F. Soluble transferrin receptor is correlated with J. Am. Soc. Nephrol. 31, 456–468 (2020).
erythropoietin sensitivity in dialysis patients. Clin. Nephrol. 52, 246–252 (1999). 129. McDonagh, T. A. et al. 2021 ESC Guidelines for the diagnosis and treatment of acute
98. Demir, A., Yarali, N., Fisgin, T., Duru, F. & Kara, A. Serum transferrin receptor levels in and chronic heart failure: developed by the Task Force for the diagnosis and treatment
beta-thalassemia trait. J. Trop. Pediatr. 50, 369–371 (2004). of acute and chronic heart failure of the European Society of Cardiology (ESC) with the
99. Birkeland, K. I. et al. Effect of rhEPO administration on serum levels of sTfR and cycling special contribution of the Heart Failure Association (HFA) of the ESC. Eur. Heart J. 42,
performance. Med. Sci. Sports Exerc. 32, 1238–1243 (2000). 3599–3726 (2021).
100. Pasricha, S. R., Rooney, P. & Schneider, H. Soluble transferrin receptor and depth of 130. Snook, J. et al. British Society of Gastroenterology guidelines for the management of iron
bone marrow suppression following high dose chemotherapy. Support. Care Cancer 17, deficiency anaemia in adults. Gut 70, 2030–2051 (2021).
847–850 (2009). 131. Yancy, C. W. et al. 2017 ACC/AHA/HFSA focused update of the 2013 ACCF/AHA Guideline
101. Gupta, S., Uppal, B. & Pawar, B. Is soluble transferrin receptor a good marker of iron for the Management of Heart Failure: a report of the American College of Cardiology/
deficiency anemia in chronic kidney disease patients? Indian J. Nephrol. 19, 96–100 American Heart Association Task Force on Clinical Practice Guidelines and the Heart
(2009). Failure Society of America. Circulation 136, e137–e161 (2017).

Nature Reviews Cardiology


Review article

132. Fletcher, A., Forbes, A., Svenson, N. & Wayne Thomas, D.; A British Society for Haematology 166. Gertler, C. et al. Magnetic resonance imaging of organ iron before and after correction of
Good Practice Paper. Guideline for the laboratory diagnosis of iron deficiency in adults iron deficiency in patients with heart failure. Esc. Heart Fail. 10, 1847–1859 (2023).
(excluding pregnancy) and children. Br. J. Haematol. 196, 523–529 (2022). 167. Maeder, M. T., Khammy, O., dos Remedios, C. & Kaye, D. M. Myocardial and systemic iron
133. Moscheo, C. et al. New insights into iron deficiency anemia in children: a practical review. depletion in heart failure implications for anemia accompanying heart failure. J. Am. Coll.
Metabolites 12, 289 (2022). Cardiol. 58, 474–480 (2011).
134. Ghio, A. J. & Hilborn, E. D. Indices of iron homeostasis correlate with airway obstruction in 168. Melenovsky, V. et al. Myocardial iron content and mitochondrial function in human heart
an NHANES III cohort. Int. J. Chron. Obstruct. Pulmon. Dis. 12, 2075–2084 (2017). failure: a direct tissue analysis. Eur. J. Heart Fail. 19, 522–530 (2017).
135. Benyamin, B. et al. Variants in TF and HFE explain approximately 40% of genetic variation 169. Zhang, H. et al. Myocardial iron deficiency and mitochondrial dysfunction in advanced
in serum-transferrin levels. Am. J. Hum. Genet. 84, 60–65 (2009). heart failure in humans. J. Am. Heart Assoc. 11, e022853 (2022).
136. Tanaka, T. et al. A genome-wide association analysis of serum iron concentrations. Blood 170. Chung, B. et al. Pre-emptive iron supplementation prevents myocardial iron deficiency
115, 94–96 (2010). and attenuates adverse remodelling after myocardial infarction. Cardiovasc. Res. 119,
137. Masini, G. et al. Criteria for iron deficiency in patients with heart failure. J. Am. Coll. Cardiol. 1969–1980 (2023).
79, 341–351 (2022). 171. Cabrera, C. et al. Relationship between iron deficiency and expression of genes involved
138. Harrison, A. V., Lorenzo, F. R. & McClain, D. A. Iron and the pathophysiology of diabetes. in iron metabolism in human myocardium and skeletal muscle. Int. J. Cardiol. 379, 82–88
Annu. Rev. Physiol. 85, 339–362 (2023). (2023).
139. Packer, M. Potential interactions when prescribing SGLT2 inhibitors and intravenous iron 172. Xu, W. et al. Lethal cardiomyopathy in mice lacking transferrin receptor in the heart.
in combination in heart failure. JACC Heart Fail. 11, 106–114 (2023). Cell Rep. 13, 533–545 (2015).
140. Sirbu, O. et al. The influence of cardiovascular medications on iron metabolism in 173. Gevaert, A. B. et al. Iron deficiency impacts diastolic function, aerobic exercise capacity,
patients with heart failure. Medicina 55, 329 (2019). and patient phenotyping in heart failure with preserved ejection fraction: a subanalysis
141. Vlahakos, V. et al. The paradigm shift from polycythemia to anemia in COPD: the critical of the OptimEx-Clin study. Front. Physiol. 12, 757268 (2021).
role of the renin-angiotensin system inhibitors. Expert Rev. Respir. Med. 16, 391–398 (2022). 174. Amaechi, U. M. et al. Clinical and echocardiographic correlates of iron status in chronic
142. Docherty, K. F. et al. Iron deficiency in heart failure and effect of dapagliflozin: findings heart failure patients: a cross-sectional descriptive study. Cureus 15, e39998 (2023).
from DAPA-HF. Circulation 146, 980–994 (2022). 175. Blayney, L., Bailey-Wood, R., Jacobs, A., Henderson, A. & Muir, J. The effects of
143. Zhao, B. et al. The relationship and clinical significance of Hepc-20, LpPLA2, PTX3, and iron deficiency on the respiratory function and cytochrome content of rat heart
acute myocardial infarction in patients with varying degrees of coronary artery disease. mitochondria. Circ. Res. 39, 744–748 (1976).
Altern. Ther. Health Med. 29, 316–321 (2023). 176. Watson, W. D. et al. Retained metabolic flexibility of the failing human heart. Circulation
144. George, M. J. et al. Novel insights into the effects of interleukin 6 antagonism in 148, 109–123 (2023).
non-ST-segment-elevation myocardial infarction employing the SOMAscan proteomics 177. Ross, K. L. & Eisenstein, R. S. Iron deficiency decreases mitochondrial aconitase
platform. J. Am. Heart Assoc. 9, e015628 (2020). abundance and citrate concentration without affecting tricarboxylic acid cycle capacity
145. Bulluck, H. et al. Residual myocardial iron following intramyocardial hemorrhage during in rat liver. J. Nutr. 132, 643–651 (2002).
the convalescent phase of reperfused ST-segment-elevation myocardial infarction and 178. Fischer, C. et al. Mitochondrial respiration in response to iron deficiency anemia:
adverse left ventricular remodeling. Circ. Cardiovasc. Imaging 9, e004940 (2016). comparison of peripheral blood mononuclear cells and liver. Metabolites 12, 270
146. Cleland, J. G. et al. Prevalence and outcomes of anemia and hematinic deficiencies in (2022).
patients with chronic heart failure. JAMA Cardiol. 1, 539–547 (2016). 179. Chung, Y. J. et al. Iron-deficiency anemia results in transcriptional and metabolic
147. Matsumoto, M. et al. Iron regulatory hormone hepcidin decreases in chronic heart failure remodeling in the heart toward a glycolytic phenotype. Front. Cardiovasc. Med. 7,
patients with anemia. Circ. J. 74, 301–306 (2010). 616920 (2020).
148. Jankowska, E. A. et al. Iron status in patients with chronic heart failure. Eur. Heart J. 34, 180. Chung, Y. J. et al. Iron-deficiency anemia reduces cardiac contraction by downregulating
827–834 (2013). RyR2 channels and suppressing SERCA pump activity. JCI Insight 4, e125618 (2019).
149. Grote Beverborg, N. et al. Definition of iron deficiency based on the gold standard of 181. Souza, J. S., Brunetto, E. L. & Nunes, M. T. Iron restriction increases myoglobin gene and
bone marrow iron staining in heart failure patients. Circ. Heart Fail. 11, e004519 (2018). protein expression in soleus muscle of rats. Acad. Bras. Cienc. 88, 2277–2290 (2016).
150. Nagai, T. et al. Prognostic significance of endogenous erythropoietin in long-term outcome 182. Hagler, L. et al. Influence of dietary iron deficiency on hemoglobin, myoglobin, their
of patients with acute decompensated heart failure. Eur. J. Heart Fail. 18, 803–813 (2016). respective reductases, and skeletal muscle mitochondrial respiration. Am. J. Clin. Nutr.
151. Carrilho, P. Intravenous iron in heart failure and chronic kidney disease. Nefrologia 41, 34, 2169–2177 (1981).
403–411 (2021). 183. Jankowska, E. A. et al. Iron deficiency predicts impaired exercise capacity in patients with
152. Xiao, L. et al. Macrophage iron retention aggravates atherosclerosis: evidence for the systolic chronic heart failure. J. Card. Fail. 17, 899–906 (2011).
role of autocrine formation of hepcidin in plaque macrophages. Biochim. Biophys. Acta 184. Tkaczyszyn, M. et al. Iron status, catabolic/anabolic balance, and skeletal muscle
Mol. Cell Biol. Lipids 1865, 158531 (2020). performance in men with heart failure with reduced ejection fraction. Cardiol. J. 28,
153. Sullivan, J. L. Iron and the sex difference in heart disease risk. Lancet 1, 1293–1294 (1981). 391–401 (2021).
154. Ekblom, K. et al. Iron stores and HFE genotypes are not related to increased risk of 185. Leermakers, P. A. et al. Iron deficiency-induced loss of skeletal muscle mitochondrial proteins
first-time myocardial infarction: a prospective nested case-referent study. Int. J. Cardiol. and respiratory capacity; the role of mitophagy and secretion of mitochondria-containing
150, 169–172 (2011). vesicles. FASEB J. 34, 6703–6717 (2020).
155. Kautz, L. et al. Testing the iron hypothesis in a mouse model of atherosclerosis. Cell Rep. 186. Perez-Peiro, M. et al. Iron depletion in systemic and muscle compartments defines
5, 1436–1442 (2013). a specific phenotype of severe COPD in female and male patients: implications in
156. Gutierrez-Bedmar, M. et al. Low serum iron levels and risk of cardiovascular disease in exercise tolerance. Nutrients 14, 3929 (2022).
high risk elderly population: nested case-control study in the PREvencion con DIeta 187. Finch, C. A. et al. Iron deficiency in the rat. Physiological and biochemical studies of
MEDiterranea (PREDIMED) trial. Clin. Nutr. 40, 496–504 (2021). muscle dysfunction. J. Clin. Invest. 58, 447–453 (1976).
157. Morrison, H. I., Semenciw, R. M., Mao, Y. & Wigle, D. T. Serum iron and risk of fatal acute 188. Beard, J., Green, W., Miller, L. & Finch, C. Effect of iron-deficiency anemia on
myocardial infarction. Epidemiology 5, 243–246 (1994). hormone levels and thermoregulation during cold exposure. Am. J. Physiol. 247,
158. Marniemi, J. et al. Dietary and serum vitamins and minerals as predictors of myocardial R114–R119 (1984).
infarction and stroke in elderly subjects. Nutr. Metab. Cardiovasc. Dis. 15, 188–197 (2005). 189. McKay, R. H., Higuchi, D. A., Winder, W. W., Fell, R. D. & Brown, E. B. Tissue effects of iron
159. Liao, Y., Cooper, R. S. & McGee, D. L. Iron status and coronary heart disease: negative deficiency in the rat. Biochim. Biophys. Acta 757, 352–358 (1983).
findings from the NHANES I epidemiologic follow-up study. Am. J. Epidemiol. 139, 190. Iversen, K. K. et al. Chronic obstructive pulmonary disease in patients admitted with
704–712 (1994). heart failure. J. Intern. Med. 264, 361–369 (2008).
160. Nordestgaard, B. G. et al. Risk factors for near-term myocardial infarction in apparently 191. Mooney, L. et al. Impact of chronic obstructive pulmonary disease in patients with heart
healthy men and women. Clin. Chem. 56, 559–567 (2010). failure with preserved ejection fraction: insights from PARAGON-HF. J. Am. Heart Assoc.
161. Hunnicutt, J., He, K. & Xun, P. Dietary iron intake and body iron stores are associated with 10, e021494 (2021).
risk of coronary heart disease in a meta-analysis of prospective cohort studies. J. Nutr. 192. Nickol, A. H. et al. A cross-sectional study of the prevalence and associations of iron
144, 359–366 (2014). deficiency in a cohort of patients with chronic obstructive pulmonary disease. BMJ Open
162. Zeller, T. et al. Prognostic value of iron-homeostasis regulating peptide hepcidin in coronary 5, e007911 (2015).
heart disease-evidence from the large atherogene study. Biomolecules 8, 43 (2018). 193. Silverberg, D. S. et al. Anemia and iron deficiency in COPD patients: prevalence and
163. Jankowska, E. A. et al. Iron deficiency defined as depleted iron stores accompanied by the effects of correction of the anemia with erythropoiesis stimulating agents and
unmet cellular iron requirements identifies patients at the highest risk of death after an intravenous iron. BMC Pulm. Med. 14, 24 (2014).
episode of acute heart failure. Eur. Heart J. 35, 2468–2476 (2014). 194. Cloonan, S. M. et al. The “Iron”-y of iron overload and iron deficiency in chronic
164. Morkedal, B., Laugsand, L. E., Romundstad, P. R. & Vatten, L. J. Mortality from ischaemic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 196, 1103–1112 (2017).
heart disease: sex-specific effects of transferrin saturation, serum iron, and total iron 195. Perez, E. et al. Hepcidin is essential for alveolar macrophage function and is disrupted
binding capacity. The HUNT study. Eur. J. Cardiovasc. Prev. Rehabil. 18, 687–694 (2011). by smoke in a murine chronic obstructive pulmonary disease model. J. Immunol. 205,
165. Pereira, G. A. R., Foppa, M., Eifer, D. A. & Beck-da-Silva, L. Myocardial iron content by 2489–2498 (2020).
T2 star cardiac magnetic resonance and serum markers of iron metabolism in patients 196. Santer, P. et al. Intravenous iron and chronic obstructive pulmonary disease:
with heart failure. J. Cardiovasc. Med. 23, e33–e35 (2022). a randomised controlled trial. BMJ Open Respir. Res. 7, e000577 (2020).

Nature Reviews Cardiology


Review article

197. Mumby, S., Saito, J., Adcock, I. M., Chung, K. F. & Quinlan, G. J. Decreased breath excretion 230. Lewis, G. D. et al. Effect of oral iron repletion on exercise capacity in patients with heart
of redox active iron in COPD: a protective failure? Eur. Respir. J. 47, 1267–1270 (2016). failure with reduced ejection fraction and iron deficiency: the IRONOUT HF randomized
198. Zhang, J. et al. Secondary polycythemia in chronic obstructive pulmonary disease: clinical trial. JAMA 317, 1958–1966 (2017).
prevalence and risk factors. BMC Pulm. Med. 21, 235 (2021). 231. Ambrosy, A. P. et al. Identifying responders to oral iron supplementation in heart
199. Shah, A. M. et al. Cardiac structure and function in heart failure with preserved ejection failure with a reduced ejection fraction: a post-hoc analysis of the IRONOUT-HF trial.
fraction: baseline findings from the echocardiographic study of the Treatment of Preserved J. Cardiovasc. Med. 20, 223–225 (2019).
Cardiac Function Heart Failure with an Aldosterone Antagonist trial. Circ. Heart Fail. 7, 232. Zdravkovic, S. C. et al. Effects of 6-months of oral ferrous and ferric supplement therapy
104–115 (2014). in patients who were hospitalized for decompensated chronic heart failure. J. Int. Med.
200. Lam, C. S. et al. Pulmonary hypertension in heart failure with preserved ejection fraction: Res. 47, 3179–3189 (2019).
a community-based study. J. Am. Coll. Cardiol. 53, 1119–1126 (2009). 233. Karavidas, A. et al. Oral sucrosomial iron improves exercise capacity and quality of life
201. Leung, C. C., Moondra, V., Catherwood, E. & Andrus, B. W. Prevalence and risk factors in heart failure with reduced ejection fraction and iron deficiency: a non-randomized,
of pulmonary hypertension in patients with elevated pulmonary venous pressure and open-label, proof-of-concept study. Eur. J. Heart Fail. 23, 593–597 (2021).
preserved ejection fraction. Am. J. Cardiol. 106, 284–286 (2010). 234. Suryani, L. D. et al. Oral ferrous sulphate improves functional capacity on heart failure
202. Chaouat, A., Naeije, R. & Weitzenblum, E. Pulmonary hypertension in COPD. Eur. Respir. J. patients with iron deficiency anemia. Glob. Heart 17, 81 (2022).
32, 1371–1385 (2008). 235. Garrido-Martin, P. et al. The effect of intravenous and oral iron administration on
203. Vonk-Noordegraaf, A. et al. Right heart adaptation to pulmonary arterial hypertension: perioperative anaemia and transfusion requirements in patients undergoing elective
physiology and pathobiology. J. Am. Coll. Cardiol. 62, D22–D33 (2013). cardiac surgery: a randomized clinical trial. Interact. Cardiovasc. Thorac. Surg. 15,
204. Oswald-Mammosser, M. et al. Prognostic factors in COPD patients receiving long-term 1013–1018 (2012).
oxygen therapy. Importance of pulmonary artery pressure. Chest 107, 1193–1198 (1995). 236. Dong, Z. et al. Comparison of oral and parenteral iron administration on iron homeostasis,
205. van Empel, V. P., Lee, J., Williams, T. J. & Kaye, D. M. Iron deficiency in patients with oxidative and immune status in anemic neonatal pigs. Biol. Trace Elem. Res. 195, 117–124
idiopathic pulmonary arterial hypertension. Heart Lung Circ. 23, 287–292 (2014). (2020).
206. Rhodes, C. J. et al. Iron deficiency and raised hepcidin in idiopathic pulmonary arterial 237. Funk, F., Fluhmann, B. & Barton, A. E. Criticality of surface characteristics of intravenous
hypertension: clinical prevalence, outcomes, and mechanistic insights. J. Am. Coll. iron-carbohydrate nanoparticle complexes: implications for pharmacokinetics and
Cardiol. 58, 300–309 (2011). pharmacodynamics. Int. J. Mol. Sci. 23, 2140 (2022).
207. Martens, P. et al. Iron deficiency in pulmonary vascular disease: pathophysiological and 238. MacDonald, B. J., Virani, S. A., Zieroth, S. & Turgeon, R. Heart failure management in
clinical implications. Eur. Heart J. 44, 1979–1991 (2023). 2023: a pharmacotherapy- and lifestyle-focused comparison of current international
208. Saluja, P. et al. Influence of iron deficiency on clinical and haemodynamic parameters in guidelines. CJC Open 5, 629–640 (2023).
pulmonary arterial hypertension cohorts. Heart Lung Circ. 31, 1594–1603 (2022). 239. Pezel, T. et al. Diagnosis and treatment of iron deficiency in heart failure: OFICSel study
209. Bart, N. K. et al. Elevation of iron storage in humans attenuates the pulmonary vascular by the French Heart Failure Working Group. ESC Heart Fail. 8, 1509–1521 (2021).
response to hypoxia. J. Appl. Physiol. 121, 537–544 (2016). 240. Becher, P. M. et al. Phenotyping heart failure patients for iron deficiency and use of
210. Talbot, N. P. et al. Contrasting effects of ascorbate and iron on the pulmonary vascular intravenous iron therapy: data from the Swedish Heart Failure Registry. Eur. J. Heart Fail.
response to hypoxia in humans. Physiol. Rep. 2, e12220 (2014). 23, 1844–1854 (2021).
211. Smith, T. G. et al. The increase in pulmonary arterial pressure caused by hypoxia depends 241. Henry, J. A. et al. 139 Cost effectiveness of IV iron in patients admitted with heart failure.
on iron status. J. Physiol. 586, 5999–6005 (2008). Heart 109, A161–A162 (2023).
212. Balanos, G. M., Dorrington, K. L. & Robbins, P. A. Desferrioxamine elevates pulmonary 242. Gonzalez-Costello, J. et al. Use of intravenous iron in patients with iron deficiency and
vascular resistance in humans: potential for involvement of HIF-1. J. Appl. Physiol. 92, chronic heart failure: real-world evidence. Eur. J. Intern. Med. 80, 91–98 (2020).
2501–2507 (2002). 243. Toblli, J. E., Lombrana, A., Duarte, P. & Di Gennaro, F. Intravenous iron reduces NT-pro-brain
213. Frise, M. C. et al. Clinical iron deficiency disturbs normal human responses to hypoxia. natriuretic peptide in anemic patients with chronic heart failure and renal insufficiency.
J. Clin. Invest. 126, 2139–2150 (2016). J. Am. Coll. Cardiol. 50, 1657–1665 (2007).
214. Howard, L. et al. Supplementation with iron in pulmonary arterial hypertension. 244. Okonko, D. O. et al. Effect of intravenous iron sucrose on exercise tolerance in anemic
Two randomized crossover trials. Ann. Am. Thorac. Soc. 18, 981–988 (2021). and nonanemic patients with symptomatic chronic heart failure and iron deficiency
215. Cancelo-Hidalgo, M. J. et al. Tolerability of different oral iron supplements: a systematic FERRIC-HF: a randomized, controlled, observer-blinded trial. J. Am. Coll. Cardiol. 51,
review. Curr. Med. Res. Opin. 29, 291–303 (2013). 103–112 (2008).
216. Low, M. S., Speedy, J., Styles, C. E., De-Regil, L. M. & Pasricha, S. R. Daily iron supplementation 245. Anker, S. D. et al. Ferric carboxymaltose in patients with heart failure and iron deficiency.
for improving anaemia, iron status and health in menstruating women. Cochrane Database N. Engl. J. Med. 361, 2436–2448 (2009).
Syst. Rev. 4, CD009747 (2016). 246. Ponikowski, P. et al. The impact of intravenous ferric carboxymaltose on renal function:
217. Schumann, K., Solomons, N. W., Orozco, M., Romero-Abal, M. E. & Weiss, G. Differences in an analysis of the FAIR-HF study. Eur. J. Heart Fail. 17, 329–339 (2015).
circulating non-transferrin-bound iron after oral administration of ferrous sulfate, sodium 247. Ponikowski, P. et al. Beneficial effects of long-term intravenous iron therapy with ferric
iron EDTA, or iron polymaltose in women with marginal iron stores. Food Nutr. Bull. 34, carboxymaltose in patients with symptomatic heart failure and iron deficiency. Eur. Heart
185–193 (2013). J. 36, 657–668 (2015).
218. Gomez-Ramirez, S., Brilli, E., Tarantino, G. & Munoz, M. Sucrosomial® iron: a new 248. van Veldhuisen, D. J. et al. Effect of ferric carboxymaltose on exercise capacity in patients
generation iron for improving oral supplementation. Pharmaceuticals 11, 97 (2018). with chronic heart failure and iron deficiency. Circulation 136, 1374–1383 (2017).
219. Asperti, M. et al. Sucrosomial((R)) iron supplementation in mice: effects on blood 249. Anker, S. D. et al. Effects of ferric carboxymaltose on hospitalisations and mortality rates
parameters, hepcidin, and inflammation. Nutrients 10, 1349 (2018). in iron-deficient heart failure patients: an individual patient data meta-analysis. Eur. J.
220. Asperti, M. et al. Iron distribution in different tissues of homozygous Mask (msk/msk) Heart Fail. 20, 125–133 (2018).
mice and the effects of oral iron treatments. Am. J. Hematol. 96, 1253–1263 (2021). 250. Dhoot, S. et al. Effect of ferric-carboxy maltose on oxygen kinetics and functional status
221. Barni, S. 5th International multidisciplinary course on iron anemia, 31st March–1 April in heart failure patients with iron deficiency. Future Sci. OA 6, FSO467 (2020).
2017, Florence, Italy. Expert Rev. Hematol. 10, 1–40 (2017). 251. Ponikowski, P. et al. Ferric carboxymaltose for iron deficiency at discharge after acute
222. Ucan, A., Kaya, Z. I., Yilmaz, E. O., Vasi, I. & Ozgeyik, M. O. Comparing therapeutic heart failure: a multicentre, double-blind, randomised, controlled trial. Lancet 396,
effects of alternate day versus daily oral iron in women with iron deficiency anemia: 1895–1904 (2020).
a retrospective cohort study. Medicine 102, e34421 (2023). 252. Kalra, P. R. et al. Intravenous ferric derisomaltose in patients with heart failure and iron
223. Schaap, C. C. et al. Diurnal rhythm rather than dietary iron mediates daily hepcidin deficiency in the UK (IRONMAN): an investigator-initiated, prospective, randomised,
variations. Clin. Chem. 59, 527–535 (2013). open-label, blinded-endpoint trial. Lancet 400, 2199–2209 (2022).
224. Troutt, J. S. et al. Circulating human hepcidin-25 concentrations display a diurnal rhythm, 253. Mollace, A. et al. Effect of ferric carboxymaltose supplementation in patients with heart
increase with prolonged fasting, and are reduced by growth hormone administration. failure with preserved ejection fraction: role of attenuated oxidative stress and improved
Clin. Chem. 58, 1225–1232 (2012). endothelial function. Nutrients 14, 5057 (2022).
225. Kreutz, J. M., Heynen, L. & Vreugdenhil, A. C. E. Nutrient deficiencies in children with 254. Graham, F. J. et al. Intravenous iron in patients with heart failure and iron deficiency:
celiac disease during long term follow-up. Clin. Nutr. 42, 1175–1180 (2023). an updated meta-analysis. Eur. J. Heart Fail. 25, 528–537 (2023).
226. Stefanelli, G., Viscido, A., Longo, S., Magistroni, M. & Latella, G. Persistent iron 255. Mentz, R. J. et al. Ferric carboxymaltose in heart failure with iron deficiency. N. Engl. J. Med.
deficiency anemia in patients with celiac disease despite a gluten-free diet. Nutrients 389, 975–986 (2023).
12, 2176 (2020). 256. Kurz, K. et al. Anaemia, iron status, and gender predict the outcome in patients with
227. Crosby, L., Palarski, V. A., Cottington, E. & Cmolik, B. Iron supplementation for chronic heart failure. Esc. Heart Fail. 7, 1880–1890 (2020).
acute blood loss anemia after coronary artery bypass surgery: a randomized, 257. Campodonico, J. et al. Prognostic role of transferrin saturation in heart failure patients.
placebo-controlled study. Heart Lung 23, 493–499 (1994). Eur. J. Prev. Cardiol. 28, 1639–1646 (2021).
228. Beck-da-Silva, L. et al. IRON-HF study: a randomized trial to assess the effects of iron in 258. Brautaset Englund, K. V. et al. Iron homeostasis in heart transplant recipients randomized
heart failure patients with anemia. Int. J. Cardiol. 168, 3439–3442 (2013). to ferric derisomaltose or placebo. Clin. Transpl. 36, e14695 (2022).
229. Clevenger, B. et al. Systematic review and meta-analysis of iron therapy in anaemic 259. Brautaset Englund, K. V. et al. Intravenous iron supplement for iron deficiency in cardiac
adults without chronic kidney disease: updated and abridged Cochrane review. transplant recipients (IronIC): a randomized clinical trial. J. Heart Lung Transpl. 40,
Eur. J. Heart Fail. 18, 774–785 (2016). 359–367 (2021).

Nature Reviews Cardiology


Review article

260. Martens, P. et al. The effect of intravenous ferric carboxymaltose on cardiac reverse 288. Wang, T., Cheng, J. & Wang, Y. Genetic support of a causal relationship between iron
remodelling following cardiac resynchronization therapy-the IRON-CRT trial. Eur. Heart J. status and atrial fibrillation: a Mendelian randomization study. Genes Nutr. 17, 8 (2022).
42, 4905–4914 (2021). 289. Zacharski, L. R. et al. Remission of paroxysmal atrial fibrillation with iron reduction in
261. Mirdamadi, A., Arefeh, A., Garakyaraghi, M. & Pourmoghadas, A. Beneficial effects of the haemophilia A. Haemophilia 16, 726–730 (2010).
treatment of iron deficiency on clinical condition, left ventricular function, and quality of 290. Jackson, I. et al. Retrospective analyses of factors influencing arrhythmias and the
life in patients with chronic heart failure. Acta Biomed. 89, 214–218 (2018). impact of arrhythmias on inpatient outcomes among hospitalized patients with
262. Toblli, J. E., Di Gennaro, F. & Rivas, C. Changes in echocardiographic parameters in iron hemochromatosis. Int. J. Cardiol. 352, 56–60 (2022).
deficiency patients with heart failure and chronic kidney disease treated with intravenous 291. Abou Yassine, A. et al. The evolution of iron-related comorbidities and hospitalization in
iron. Heart Lung Circ. 24, 686–695 (2015). patients with hemochromatosis: an analysis of the nationwide inpatient sample. Blood
263. Nunez, J. et al. Noninvasive imaging estimation of myocardial iron repletion following Sci. 5, 131–135 (2023).
administration of intravenous iron: the myocardial-IRON trial. J. Am. Heart Assoc. 9, 292. Dai, C. et al. Inhibition of ferroptosis reduces susceptibility to frequent excessive alcohol
e014254 (2020). consumption-induced atrial fibrillation. Toxicology 465, 153055 (2022).
264. Charles-Edwards, G. et al. Effect of iron isomaltoside on skeletal muscle energetics 293. Rose, R. A. et al. Iron overload decreases CaV1.3-dependent L-type Ca2+ currents leading
in patients with chronic heart failure and iron deficiency. Circulation 139, 2386–2398 to bradycardia, altered electrical conduction, and atrial fibrillation. Circ. Arrhythm.
(2019). Electrophysiol. 4, 733–742 (2011).
265. Caravita, S. et al. Intravenous iron therapy improves the hypercapnic ventilatory response 294. Kumfu, S., Chattipakorn, S., Chinda, K., Fucharoen, S. & Chattipakorn, N. T-type calcium
and sleep disordered breathing in chronic heart failure. Eur. J. Heart Fail. 24, 1940–1949 channel blockade improves survival and cardiovascular function in thalassemic mice.
(2022). Eur. J. Haematol. 88, 535–548 (2012).
266. Okonko, D. O. et al. Effect of ferric carboxymaltose on calculated plasma volume status 295. Tu, S. J. et al. Rationale and design of the IRON-AF study: a double-blind, randomised,
and clinical congestion: a FAIR-HF substudy. ESC Heart Fail. 6, 621–628 (2019). placebo-controlled study to assess the effect of intravenous ferric carboxymaltose in
267. Abdula, G. et al. Ferric carboxymaltose has a higher distribution into myocardium than patients with atrial fibrillation and iron deficiency. BMJ Open. 11, e047642 (2021).
gadobutrol — a quantitative T1 mapping study in healthy volunteers. Preprint at medRxiv 296. Galetti, V. et al. Threshold ferritin and hepcidin concentrations indicating early iron
https://doi.org/10.1101/2023.03.01.23285660 (2023). deficiency in young women based on upregulation of iron absorption. EClinicalMedicine
268. Span, K. et al. The use of magnetic resonance imaging for non-invasive assessment of 39, 101052 (2021).
venofer® biodistribution in rats. Pharm. Res. 35, 88 (2018). 297. Jankowska, E. A. et al. Bone marrow iron depletion is common in patients with coronary
269. Rostoker, G. et al. Differential pharmacokinetics of liver tropism for iron sucrose, ferric artery disease. Int. J. Cardiol. 182, 517–522 (2015).
carboxymaltose, and iron isomaltoside: a clue to their safety for dialysis patients. 298. Everett, B. M. et al. Anti-inflammatory therapy with canakinumab for the prevention of
Pharmaceutics 14, 1408 (2022). hospitalization for heart failure. Circulation 139, 1289–1299 (2019).
270. De Souza, L. V. et al. Comparative analysis of oral and intravenous iron therapy in rat 299. Ridker, P. M. et al. Anti-inflammatory therapy with canakinumab for atherosclerotic
models of inflammatory anemia and iron deficiency. Haematologica 108, 135–149 (2023). disease. N. Engl. J. Med. 377, 1119–1131 (2017).
271. Bailie, G. R. et al. Data from the Dialysis Outcomes and Practice Patterns Study validate 300. Breenfeldt Andersen, A. et al. Effects of altitude and recombinant human erythropoietin
an association between high intravenous iron doses and mortality. Kidney Int. 87, 162–168 on iron metabolism: a randomized controlled trial. Am. J. Physiol. Regul. Integr. Comp.
(2015). Physiol. 321, R152–R161 (2021).
272. Kalantar-Zadeh, K., Regidor, D. L., McAllister, C. J., Michael, B. & Warnock, D. G. 301. Robach, P. et al. Induction of erythroferrone in healthy humans by micro-dose recombinant
Time-dependent associations between iron and mortality in hemodialysis patients. erythropoietin or high-altitude exposure. Haematologica 106, 384–390 (2021).
J. Am. Soc. Nephrol. 16, 3070–3080 (2005). 302. Palazzuoli, A. et al. Erythropoietin improves anemia exercise tolerance and renal
273. Oudit, G. Y. et al. L-type Ca2+ channels provide a major pathway for iron entry into function and reduces B-type natriuretic peptide and hospitalization in patients with
cardiomyocytes in iron-overload cardiomyopathy. Nat. Med. 9, 1187–1194 (2003). heart failure and anemia. Am. Heart J. 152, 1096.e9-15 (2006).
274. Udani, K. et al. Cardiovascular manifestations in hospitalized patients with 303. Silverberg, D. S. et al. The effect of correction of mild anemia in severe, resistant
hemochromatosis in the United States. Int. J. Cardiol. 342, 117–124 (2021). congestive heart failure using subcutaneous erythropoietin and intravenous iron:
275. Schouten, B. J., Hunt, P. J., Livesey, J. H., Frampton, C. M. & Soule, S. G. FGF23 elevation a randomized controlled study. J. Am. Coll. Cardiol. 37, 1775–1780 (2001).
and hypophosphatemia after intravenous iron polymaltose: a prospective study. J. Clin. 304. Singh, A. K. et al. Correction of anemia with epoetin alfa in chronic kidney disease.
Endocrinol. Metab. 94, 2332–2337 (2009). N. Engl. J. Med. 355, 2085–2098 (2006).
276. Yang, F., Wang, W., Zhang, Y., Nong, J. & Zhang, L. Effects of ferroptosis in myocardial 305. Akizawa, T. et al. Iron regulation by molidustat, a daily oral hypoxia-inducible factor prolyl
ischemia/reperfusion model of rat and its association with Sestrin 1. Adv. Clin. Exp. Med. hydroxylase inhibitor, in patients with chronic kidney disease. Nephron 143, 243–254 (2019).
32, 219–231 (2023). 306. Renders, L. et al. First-in-human phase I studies of PRS-080#22, a hepcidin antagonist,
277. Miyamoto, H. D. et al. Iron overload via heme degradation in the endoplasmic reticulum in healthy volunteers and patients with chronic kidney disease undergoing hemodialysis.
triggers ferroptosis in myocardial ischemia-reperfusion injury. JACC Basic Transl. Sci. 7, PLoS One 14, e0212023 (2019).
800–819 (2022). 307. Hohlbaum, A. M. et al. Sustained plasma hepcidin suppression and iron elevation by
278. Cosentino, N. et al. Iron deficiency in patients with ST-segment elevation myocardial anticalin-derived hepcidin antagonist in cynomolgus monkey. Br. J. Pharmacol. 175,
infarction undergoing primary percutaneous coronary intervention. Int. J. Cardiol. 300, 1054–1065 (2018).
14–19 (2020).
279. Roghi, A. et al. Role of non-transferrin-bound iron in the pathogenesis of cardiotoxicity Acknowledgements
in patients with ST-elevation myocardial infarction assessed by cardiac magnetic S.L.-L. is funded by MR/V009567/1/ from the Medical Research Council, UK, and by HSR00031
resonance imaging. Int. J. Cardiol. 199, 326–332 (2015). from the British Heart Foundation, UK.
280. Paraskevaidis, I. A. et al. Deferoxamine infusion during coronary artery bypass grafting
ameliorates lipid peroxidation and protects the myocardium against reperfusion injury: Author contributions
immediate and long-term significance. Eur. Heart J. 26, 263–270 (2005). S.L.-L. researched data for the article and wrote the manuscript. Both authors reviewed/edited
281. Haddad, S. et al. Iron-regulatory proteins secure iron availability in cardiomyocytes to the manuscript before submission.
prevent heart failure. Eur. Heart J. 38, 362–372 (2017).
282. Florian, A. et al. Positive effect of intravenous iron-oxide administration on left ventricular Competing interests
remodelling in patients with acute ST-elevation myocardial infarction — a cardiovascular S.L.-L. reports receipt of previous research funding from Vifor Pharma, personal honoraria for
magnetic resonance (CMR) study. Int. J. Cardiol. 173, 184–189 (2014). a lecture from Pharmacosmos, and consultancy fees from Disc Medicine and ScholarRock.
283. Wischmann, P. et al. Safety and efficacy of iron supplementation after myocardial infarction J.G.F.C. reports personal honoraria for lectures and advisory boards from Amgen,
in mice with moderate blood loss anaemia. Esc. Heart Fail. 8, 5445–5455 (2021). AstraZeneca, Bayer, Novartis, Pharmacosmos, Servier and Vifor Pharma.
284. Theidel, U. et al. Budget impact of intravenous iron therapy with ferric carboxymaltose
in patients with chronic heart failure and iron deficiency in Germany. ESC Heart Fail. 4, Additional information
274–281 (2017). Peer review information Nature Reviews Cardiology thanks Gavin Oudit, Gianluigi Savarese
285. Shokri, H. & Ali, I. Intravenous iron supplementation treats anemia and reduces blood and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
transfusion requirements in patients undergoing coronary artery bypass grafting — a
prospective randomized trial. Ann. Card. Anaesth. 25, 141–147 (2022). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
286. Kim, H. H., Park, E. H., Lee, S. H., Yoo, K. J. & Youn, Y. N. Effect of preoperative administration published maps and institutional affiliations.
of intravenous ferric carboxymaltose in patients with iron deficiency anemia after off-pump
coronary artery bypass grafting: a randomized controlled trial. J. Clin. Med. 12, 1737 (2023). Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this
287. Johansson, P. I., Rasmussen, A. S. & Thomsen, L. L. Intravenous iron isomaltoside 1000 article under a publishing agreement with the author(s) or other rightsholder(s); author
(Monofer®) reduces postoperative anaemia in preoperatively non-anaemic patients self-archiving of the accepted manuscript version of this article is solely governed by the
undergoing elective or subacute coronary artery bypass graft, valve replacement terms of such publishing agreement and applicable law.
or a combination thereof: a randomized double-blind placebo-controlled clinical trial
(the PROTECT trial). Vox Sang. 109, 257–266 (2015). © Springer Nature Limited 2024

Nature Reviews Cardiology

You might also like