Int J Heart Fail. 2023 Oct;5(4):173-183. English.
Published online Oct 25, 2023.
Copyright © 2023. Korean Society of Heart Failure
Review

Iron Deficiency in Heart Failure: A Korea-Oriented Review

Ewa A. Jankowska, MD, PhD,1,2 and Piotr Ponikowski, MD, PhD1,2
    • 1Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland.
    • 2Institute of Heart Diseases, 10 University Hospital, Wroclaw, Poland.
Received June 16, 2023; Revised August 24, 2023; Accepted September 27, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Iron deficiency (ID) occurs at high frequency across the spectrum of heart failure (HF), with HF severity and race being potentially important predictors for its development. ID, irrespective of anaemia status, leads to poor outcomes in patients with HF, including exacerbated reduction in exercise capacity, poor quality of life (QoL) and increased risk of HF hospitalisation. As ID has a large public health and economic burden in Asia, and patients hospitalised with acute HF in the Asia Pacific vs. other regions commonly present with more severe clinical symptoms, there is a clear need to identify and treat ID promptly in Asian patients with HF. The biomarkers serum ferritin and transferrin saturation are used for ID diagnosis, and periodic screening is recommended in all patients with HF. The intravenous iron treatments, ferric carboxymaltose (FCM) and ferric derisomaltose, have demonstrated efficacy and tolerability in patients with acute or chronic HF and ID, with FCM shown to be cost-effective (and in some cases cost-saving). Meta-analyses support the likely benefits of intravenous FCM for improving QoL and reducing HF hospitalisation, without reducing mortality risk in patients with HF and ID. Accordingly, European Society of Cardiology guidelines recommend considering intravenous FCM for patients with symptomatic HF with left ventricular ejection fraction ≤50% who were recently hospitalised for HF and have ID. Although analyses of Asian patients with HF and ID are limited, the effects of intravenous iron would be expected to be similar to that in White populations; further clarifying studies may be of interest.

Keywords
Asia; Heart failure; Iron deficiency; Iron; Supplies; Korea

EPIDEMIOLOGY OF IRON DEFICIENCY (ID)

ID is the most prevalent micronutrient deficiency, affecting greater than one-third of the world’s population.1) It can result in microcytic anaemia, causing symptoms such as fatigue, weakness and shortness of breath.1) ID is particularly common in countries with a low and low–middle sociodemographic index.1) In 2019, the age-standardised prevalence rate of ID per 100,000 persons was greater than 14,000 globally, with rates in Asia varying from 4,724 in East Asia to 27,404 in South Asia.1)

EPIDEMIOLOGY OF ID IN HEART FAILURE (HF)

HF is a considerable health burden, affecting approximately 64.3 million people globally.2) In the Western Pacific region, HF has been found to occur at a slightly younger mean age of 67 years compared with 70 years in the USA, with an even younger mean age of 54 years observed for HF diagnosis in Southeast Asia.3) In Korea specifically, the prevalence of HF has been increasing in the last 2 decades, from 0.77% of the population reported as having HF in 2002 to 2.24% reported in 2018.4)

ID is a non-cardiovascular co-morbidity that occurs at high frequency across the whole spectrum of HF, affecting as many as 37–83% of patients with HF overall and 65–83% of patients following an acute HF episode.5, 6, 7, 8, 9) In a multi-ethnic Southeast Asian population, the odds of having ID was 3.5-fold higher in patients with HF than control patients who did not have HF.10) In this study, the prevalence of ID in the Southeast Asian population with HF (61%) exceeded that reported for European counterparts with HF (37–50%).10) In Korea, 53.8% of patients hospitalised with acute HF were reported to have ID.11)

HF severity and race have been highlighted as important predictors of ID in patients with HF. In global studies, significant, independent associations were seen between ID and New York Heart Association Functional Classification (NYHA) > II, high serum high-sensitivity C-reactive protein, and high plasma N-terminal pro-B-type natriuretic peptide (NT-proBNP).7, 12) In a Southeast Asian population, a lower left ventricular ejection fraction (LVEF), and being an inpatient (vs. an outpatient), with a higher body-mass index, and Indian (vs. Chinese) race were all associated with significantly increased odds of having ID.10) In Korea, a higher heart rate, anaemia, and use of clopidogrel were independent predictors of ID in patients hospitalised with acute HF, as was female sex.11) Although these factors can help predict patients at the highest risk of ID, periodic ID screening is recommended for all patients with HF.13)

ID is associated with poorer outcomes in patients with HF, irrespective of whether the patient has anaemia or not.7, 8, 14) In a prospective, multicentre cohort study in Korea, 53.8% of patients hospitalised with acute HF had ID, while only 34.9% had ID anaemia.11)

PATHOPHYSIOLOGY OF ID IN HF

Iron is essential for the functioning of haematopoietic and non-haematopoietic cells. Iron is required for erythropoiesis and immune responses, is present in every cell and is essential for various metabolic processes(Figure 1).15, 16, 17) Therefore, ID has critical consequences for organs in which the tissue has high energy requirements, such as the kidneys, skeletal muscle and myocardium(Figure 2).17, 18)

Figure 1
Involvement of iron in haematopoietic and non-haematopoietic metabolic pathways.15, 16, 17)
ROS = reactive oxygen species.

Figure 2
Iron deficiency impairs functioning of organs characterised by high daily energy consumption.17, 18)
HF = heart failure; LV = left ventricular.

The mechanism behind the high prevalence of ID in HF has not been fully elucidated. One hypothesis is that the mechanism underpinning ID in chronic kidney disease (inflammation leading to a cascade of hepcidin production, which triggers the migration of iron into reticuloendothelial cells where it is unavailable for metabolic processes) may also apply to the pathogenesis of ID in HF.17, 19, 20, 21) However, the inflammation-catalysed iron redistribution hypothesis remains disputed because of the limited supporting evidence for inflammation as a catalyst in ID and the contrasting finding that patients with chronic22, 23) or acute8) HF have very low levels of circulating hepcidin as opposed to high levels of hepcidin. This suggests that ID in patients with HF may instead be associated with iron store depletion.23) The reliability of hepcidin as a sole marker for ID in HF may be limited because the level of hepcidin can be influenced by various factors.24)

Iron store depletion may result from a reduction in iron consumption or iron absorption, abnormal iron loss (i.e., from gastrointestinal disorders such as peptic ulcers and colitis, or blood loss from the urinary tract) or iron deposition to stores that are inaccessible for metabolic processes.24) However, while these mechanisms are logical, their contribution to ID in HF remains unclear. More research is required to investigate the mechanisms underlying ID in HF.

DEFINING ID IN HF

Haematological assessment of iron stores within bone marrow is considered the gold standard approach to diagnosing ID.16, 24) However, the invasiveness and lack of availability of the bone aspiration procedure renders it impractical outside specialist haematology settings.24) Additionally, results from bone marrow predominantly provide insight into iron status for erythropoietic processes. Thus, biomarker identification is considered a more suitable method to diagnose ID in patients with HF.24) Serum ferritin and transferrin saturation (TSAT) are the standard biomarkers recommended in European Society of Cardiology (ESC) guidelines for diagnosing ID in patients with HF.13) Ferritin is a blood protein that stores iron and is predominantly found in reticuloendothelial cells and hepatocytes.25) Transferrin is a protein that transports iron in the blood serum; thus, the measure of TSAT is given as a percentage of transferrin binding and transporting iron. This percentage is calculated as the serum iron divided by the total iron binding capacity of transferrin, which is then multiplied by 100.17, 26, 27)

One proposed definition of ID in HF is low levels of serum ferritin (<100 ng/mL).13) However, ferritin production is increased during periods of inflammation (as seen in HF), meaning that serum ferritin alone cannot be used to definitively diagnose ID in HF.17, 27) Therefore, a secondary biomarker, TSAT, is used to provide further indication of iron depletion.17, 27) In the presence of ID, transferrin is upregulated, leading to a higher volume of circulating non-iron bound transferrin, which results in a relative TSAT decrease. Thus, a decrease in TSAT indicates ID. Therefore, the second definition of ID in HF is defined as TSAT <20% with serum ferritin 100–299 ng/mL.13) To account for the inflammation seen in HF, researchers and treating clinicians apply a high cut-off level for serum ferritin in combination with a low TSAT to diagnose ID in HF more accurately.17, 27) ESC 2021 HF guidelines and the 2022 Korean Society of Heart Failure (KSHF) Guideline for the Management of HF recommend diagnosis of ID in patients with HF if serum ferritin values are <100 ng/mL, or 100–299 ng/mL accompanied by TSAT <20%.13, 28) This definition has been used as an inclusion criterion in several clinical studies investigating the effect of intravenous (IV) iron in patients with ID and HF, which have demonstrated improved clinical outcomes.29, 30, 31) This suggests that the definition can help to identify patients with ID who are likely to benefit from iron-repletion therapy.

An alternative definition of ID has been explored using the novel iron biomarkers: hepcidin and soluble transferrin receptors.8, 22) Low serum hepcidin reflects depleted iron stores more accurately than ferritin, while high soluble transferrin receptors reflect depleted intracellular iron that is insufficient for metabolic processes.8, 22) Future studies assessing the specificity and selectivity of different combinations and cut-offs of the aforementioned ID biomarkers would be informative for their potential diagnostic application and to help guide therapy.

CLINICAL CONSEQUENCES OF ID IN HF

HF is a condition in which there is high morbidity and mortality.32) ID (irrespective of anaemia status) exacerbates the clinical symptoms of HF33) and has been associated with reduced exercise capacity,34) poor health-related quality of life (QoL),35) increased risk of non-fatal cardiovascular events (HF hospitalisation, acute coronary syndrome, severe arrythmia and stroke),14) and increased risk of all-cause death,7) as well as high healthcare costs (Figure 3).36)

Figure 3
Clinical consequences of iron deficiency in patients with heart failure.7, 14, 33, 34, 35, 36)

Patients hospitalised with acute HF in the Asia Pacific region commonly present with more severe clinical symptoms and at an earlier age than patients from other regions,37) suggesting an ‘Asian phenotype’ of aggressive disease progression.38) This highlights the large public health and economic burden of HF in Asia.10) As such, the need to identify and treat ID promptly in patients with HF in Asia may be even more important.

EXISTING DATA ON IRON REPLETION THERAPY IN PATIENTS WITH HF

While inexpensive and widely available, oral formulations of iron sulphate, gluconate, or fumarate39) are not recommended to treat iron deficiency in patients with HF13) because there is a lack of evidence for their clinical benefit.40, 41) However, there is growing evidence supporting the use of IV iron treatment for patients with HF, which is recognised in the ESC13) and the 2022 KSHF Guideline for the Management of HF.28) IV ferric carboxymaltose (FCM) and ferric derisomaltose (FDI) are iron (III) hydroxides in complex with carboxymaltose42) and derisomaltose,43) respectively. While the exact mechanism of action of iron (III) hydroxide complexes—such as FCM—is not known, studies have reported that iron released from these complexes binds to transferrin and is delivered to reticuloendothelial cells.42, 44) In clinical trials, FCM and FDI have demonstrated efficacy and tolerability in patients with ID in acute and chronic HF (results are summarised in Table 1).29, 30, 31, 45) Initial adverse effects such as nausea, hypotension and peripheral oedema were originally observed with IV iron due to the oxyhydroxide complex formulation; however, in the more recently-developed formulations, FCM and FDI, the iron is encased in a carbohydrate shell, largely averting these adverse effects.29, 30, 31, 45, 46)

Table 1
Intravenous iron in heart failure clinical trials

Iron sucrose is another type of IV iron complex, and while it is a different complex to that of FCM and FDI, it has also showcased desirable effects in patients with HF.47, 48) In the FERRIC-HF48) and IRON-HF47) trials, iron sucrose use resulted in an improvement in exercise capacity in patients with HF. However, patients can be administered a much higher iron dosage in a single administration with FCM compared with iron sucrose, which requires multiple administrations to reach the optimal dose.39)

Key trials investigating IV iron in patients with ID and HF

The FAIR-HF and CONFIRM-HF trials investigated the effects of IV FCM vs. placebo on clinical and QoL outcomes in patients with ID (with or without anaemia) and chronic HF with reduced ejection fraction (LVEF <45%).29, 30, 49) In both trials, improvements in 6-minute walk test (6MWT) distance, QoL, NYHA class, and self-reported disease activity and overall health (based on patient global assessment score), were seen with FCM vs. placebo. In CONFIRM-HF, IV FCM was also found to reduce the secondary endpoint of risk of hospitalisation for worsening HF at week 52.30) Results were found to be independent of anaemia status.29, 49)

Two pooled analyses of the FAIR-HF and CONFIRM-HF trials with increased statistical power reinforced the findings of the individual trials. In the first pooled analysis, significantly higher proportions of patients in the FCM arm experienced a clinically meaningful (≥20 m) improvement in 6MWT compared with placebo at week 12 (56.8% vs. 37.4%; odds ratio [OR], 2.156; 95% confidence interval [CI], 1.571–2.960; p<0.0001).50) Among the patients who had a significant improvement in 6MWT at week 12, >80% sustained this improvement at week 24.50) In the second pooled analysis, a significantly higher proportion of patients experienced a clinically meaningful (≥4.3-point) improvement in Kansas City Cardiomyopathy Questionnaire (KCCQ) overall summary score (OSS; representing QoL) with IV FCM than placebo at week 12 (60.5% vs. 46.6%; OR, 1.75; 95% CI, 1.26–2.44; p=0.0008).51) Thus, pooled data from FAIR-HF and CONFIRM-HF showed that FCM can improve HF clinical status, exercise capacity and QoL in patients with ID and chronic HF with reduced ejection fraction compared with placebo.50, 51)

Following on from the success of FCM in patients with ID and chronic HF, the AFFIRM-AHF clinical trial investigated the effect of IV FCM compared with placebo in patients with ID after stabilisation following an acute HF episode (when risk of rehospitalisation and mortality is high) with LVEF <50%.31) Given the lack of data on the safety of IV iron supplementation in this high-risk population, repeat dosing was only performed up to week 24 in AFFIRM-AHF, with clinical efficacy endpoints assessed at week 52. The trial narrowly missed its composite primary endpoint of total HF hospitalisations and cardiovascular death at week 52 (rate ratio [RR], 0.79; 95% CI, 0.62–1.01; p=0.059). This was potentially in part due to the impact of the coronavirus disease 2019 (COVID-19) pandemic on data collection and follow-up. A sensitivity analysis that censored patients at the first date of COVID-19 detection in their region revealed a significant effect of FCM vs. placebo on the primary endpoint (RR, 0.75; 95% CI, 0.59–0.96; p=0.024).

In terms of secondary endpoints in AFFIRM-AHF, IV FCM was associated with a significant reduction in total HF hospitalisations vs. placebo at week 52 (RR, 0.74; 95% CI, 0.58–0.94; p=0.013),31) as well as a significantly greater improvement in QoL score (evaluated using KCCQ-12 OSS and clinical summary score) as early as week 4 after the start of treatment (adjusted mean difference for OSS: 2.9, 95% CI, 0.5–5.3; p=0.018), lasting up to week 24 (adjusted mean difference for OSS: 3.0, 95% CI, 0.3–5.6; p=0.028).52) These latter findings were drivers of dominance (cost saving with additional health improvement; Switzerland, UK and USA) and high cost-effectiveness (Italy) in a pharmacoeconomic analysis of IV FCM in acute HF based on AFFIRM-AHF data,36) with no effect of FCM vs. placebo on cardiovascular death observed in the AFFIRM-AHF trial (hazard ratio [HR], 0.96; 95% CI, 0.70–1.32; p=0.81).31) Other secondary clinical endpoints, including time to first HF hospitalisation or cardiovascular death (HR, 0.80; 95% CI, 0.66–0.98; p=0.030) and days lost due to HF hospitalisations and cardiovascular death (RR, 0.67; 95% CI, 0.47–0.97; p=0.035) also significantly favoured FCM vs. placebo31); however, secondary endpoints were considered hypothesis-generating only, given the non-significant primary endpoint.

The investigator-initiated, open-label, randomised IRONMAN trial investigating another IV iron preparation, FDI, in patients with ID and new or established symptomatic HF with LVEF ≤45%, was reported in November 2022.45) This trial assessed similar outcomes to the AFFIRM-AHF trial, but in a lower-risk population and over a longer follow-up period (median of 2.7 years), with repeat dosing permitted throughout the trial.

While AFFIRM-AHF only enrolled patients during hospitalisation for an acute HF episode, IRONMAN also enrolled non-hospitalised patients with elevated plasma NT-proBNP, elevated plasma B-type natriuretic peptide or prior hospitalisation for HF within the last 6 months. Furthermore, the iron parameters indicating a patient for redosing in IRONMAN (serum ferritin <100 µg/L or <400 µg/L if TSAT <25%) were more lenient compared with prior trials.53) Despite these differences, IRONMAN data were largely in agreement with findings from the AFFIRM-AHF trial.

While the composite primary endpoint of recurrent hospitalisations for HF and cardiovascular death with IV FDI vs. usual care (excluding IV iron) was missed in IRONMAN, a numeric reduction in this endpoint was observed (RR, 0.82; 95% CI, 0.66–1.02; p=0.070). Similar to AFFIRM-AHF, the COVID-19 pandemic had a considerable impact on the IRONMAN trial, influencing the ability of patients to attend in-person visits for assessment of iron levels and repeat IV FDI dosing, as well as recruitment and retention. As in AFFIRM-AHF, when a sensitivity analysis accounting for the effects of COVID-19 in IRONMAN was performed, a significant reduction in the primary endpoint was observed with IV FDI compared with usual care (RR, 0.76; 95% CI, 0.58–1.00; p=0.047). This suggests that COVID-19 was a key, unanticipated interferent in both AFFIRM-AHF and IRONMAN statistical analysis plans, with potentially greater influence in IRONMAN due to COVID-19 restricting FDI dosing from March 2020.

The impact of COVID-19 is also reflected in many of the secondary clinical endpoints in IRONMAN, including hospitalisation for HF (RR, 0.80; 95% CI, 0.62–1.03; p=0.085) and change in 6MWT at 20 months (estimated mean difference: −35.9, 95% CI, −74.4 to 2.64; p=0.068). Only the secondary endpoint of time to first cardiovascular death or hospital admission for stroke, myocardial infarction or HF was significantly reduced in the FDI vs. usual care arm (HR, 0.83; 95% CI, 0.69–1.00; p= 0.045). Additionally, although patients treated with IV FDI had a significantly better overall Minnesota Living with HF Questionnaire (MLHFQ) score at 4 months (adjusted mean difference −3.33, 95% CI, −6.67 to 0.00; p=0.050) when compared with the usual care group, no significant difference in overall MLHFQ score was observed between the 2 groups at 20 months (adjusted mean difference −2.57, 95% CI, −6.72 to 1.59; p=0.23).The role of COVID-19 in limiting FDI redosing may potentially have affected this outcome, similar to the attenuation of the treatment effect after cessation of FCM dosing in AFFIRM-AHF.52)

Taken together, results from individual trials support the likely benefit of IV iron supplementation for reducing hospitalisation for HF and improving QoL across a spectrum of patients with acute or chronic HF and ID, with the cost-effectiveness (and in some cases cost-savings) of FCM predicted from a number of healthcare system perspectives.

IV iron in Asian populations with ID and HF

Of the 1,108 patients included in AFFIRM-AHF analyses and the 1,137 patients included in IRONMAN analyses, 4.3% and 5.8%, respectively, were of Asian race,29, 45) with analyses by race currently unavailable. No patients from Asia were included in the FAIR-HF and CONFIRM-HF analyses; however, FAIR-HF data have been used to perform cost-effectiveness and cost-utility analyses of FCM in patients with chronic HF and ID anaemia from a Korean healthcare payer perspective, based on the ratio of healthcare resource utilisation in Korea using NYHA class and NYHA improvement rates in FAIR-HF.54) In the base-case scenario, IV FCM was cost-effective vs. placebo for ID, with an incremental cost-effectiveness ratio of ₩25,010,451 ($22,192) per quality-adjusted life year.

One small pilot study, PRACTICE-ASIA-HF, of IV iron in Southeast Asian patients with ID and HF has been conducted.55) A single 1,000 mg dose of IV FCM or placebo was administered to 50 Southeast Asian patients with ID (defined as serum ferritin <300 ng/mL with TSAT <20%), who had stabilised following acute decompensated HF (regardless of LVEF) (results are summarised in Table 1).55) This study differed from other trials of IV FCM (FAIR-HF, CONFIRM-HF and AFFIRM-AHF) in several ways. To account for the average lower weight of a Southeast Asian patient with HF compared with a Caucasian patient with HF and assuming an Hb of ≥10 g/dL, a fixed dose of 1,000 mg was administered in PRACTICE-ASIA-HF, with no repeat dosing for persistent ID. This resulted in incomplete iron repletion in some patients. As PRACTICE-ASIA-HF was a pilot study, it was small (25 patients per treatment arm) and relatively short (12 weeks), which greatly limited the statistical power available to detect significant treatment effects.

PRACTICE-ASIA-HF found an increase in mean 6MWT distance from 252 m at baseline to 334 m at week 12 in the FCM arm, with continued incremental improvement throughout the 12 weeks, and from 243 m at baseline to 301 m at week 12 in the placebo arm, with a plateau after 4 weeks. However, no significant effect of FCM vs. placebo on change from baseline in 6MWT distance at 12 weeks (primary endpoint) was observed following covariate adjustment (mean difference: 0.88 m, 95% CI, −30.2 to 32.0; p=0.956). Given that significant treatment effects for the 6MWT became apparent at 24 weeks in CONFIRM-HF,30) the shorter study duration of PRACTICE-ASIA-HF may have been insufficient to capture significant improvements in exercise capacity with IV FCM.

Similarly, changes in QoL measures (secondary endpoints) did not differ significantly between treatment arms in PRACTICE-ASIA-HF. Respective mean KCCQ OSS scores at baseline and week 12 increased from 50.0 to 82.4 in the FCM arm and from 51.2 to 87.1 in the placebo arm (adjusted mean difference: −1.48, 95% CI, −8.27 to 5.31; p=0.67). Mean visual analogue scale scores increased from 6.4 at baseline to 7.7 at week 4 in the FCM arm, before decreasing slightly to 6.8 at week 12 (potentially highlighting initial benefits of iron repletion followed by fall-off of the effect due to lack of repeat dosing); in the placebo arm, scores increased from 5.7 at baseline to 6.9 at week 4, remaining stable thereafter (adjusted mean difference in change from baseline to week 12 with FCM vs. placebo: 0.26, 95% CI, −0.33 to 0.86; p=0.39). IV FCM was well tolerated in PRACTICE-ASIA-HF with no serious treatment-associated adverse events reported. Given the aforementioned limitations of PRACTICE-ASIA-HF, data from this pilot study are encouraging but insufficient to draw firm conclusions regarding the efficacy of IV FCM in Southeast Asian patients with ID following an acute HF episode; further clarifying studies in this population may be informative.

Totality of evidence

A meta-analysis investigated the effect of IV iron in 5 trials of patients with systolic HF (LVEF ≤45%) and ID.56) The FAIR-HF30) and CONFIRM-HF30) trials included in the meta-analysis investigated IV FCM vs. placebo, whereas the Toblli et al.,57) FERRIC-HF,48) and IRON-HF47) trials investigated IV iron sucrose vs. placebo. Patients in IRON-HF were also treated with oral ferrous sulphate. This meta-analysis revealed that IV iron reduced the composite endpoint of hospitalisation for cardiovascular causes or all-cause death by 66% vs. placebo (OR, 0.44; 95% CI, 0.30–0.64; p<0.0001), as well as the composite endpoint of hospitalisation for worsening HF or cardiovascular death (OR, 0.39; 95% CI, 0.24–0.63; p=0.0001). Outcomes for which individual endpoints were given as mean net effects showed a reduction in NYHA class of −0.54 classes (95% CI, −0.87 to −0.21; p=0.001), an increase in 6MWT distance of 31 m (95% CI, 18–43; p<0.0001) and an overall improvement in QoL scores with FCM vs. placebo. These findings support those from individual trials.

A second meta-analysis that extracted individual patient data from 4 double-blind, randomised controlled trials (FER-CARS-01, FAIR-HF, EFFICACY-HF and CONFIRM-HF) found that IV FCM significantly reduced the rates of recurrent cardiovascular hospitalisations and cardiovascular mortality vs. placebo in patients with systolic HF and ID (RR, 0.59; 95% CI, 0.40–0.88; p=0.009).58) However, a more recent meta-analysis that included 7 trials in patients with HF and ID suggested that a reduction in cardiovascular, rather than mortality, events with IV iron, may be the key driver of composite endpoint results including such events.59) In this meta-analysis, IV iron reduced the rate of HF hospitalisations or cardiovascular death (OR, 0.73; 95% CI, 0.59–0.90; p=0.003); however when the individual components of this composite endpoint were analysed, the rate of first HF hospitalisation remained significantly reduced (OR, 0.67; 95% CI, 0.54–0.85; p=0.0007), but the rate of cardiovascular death did not (OR, 0.89; 95% CI, 0.66–1.21; p=0.47). This finding is in agreement with the results of the AFFIRM-AHF analysis, which showed a reduction in HF hospitalisations but not cardiovascular death with FCM vs. placebo.31)

Thus, the totality of evidence suggests that IV iron should be considered in patients with ID and HF to reduce hospitalisation rates, and improve functional outcomes and QoL, without an expectation for reduced mortality risk. Indeed, recommendations surrounding the use of IV iron in patients with HF are now included in a number of regional cardiology society guidance documents: the ESC guidelines include a class IIa recommendation to consider the use of IV FCM to reduce the risk of HF hospitalisation in patients with symptomatic HF, LVEF ≤50% and ID who have been recently hospitalised for HF13); and the Asian Pacific Society of Cardiology consensus statements include a recommendation to consider IV FCM in patients with symptomatic chronic HF with reduced (moderate level of evidence; 100% consensus) or mildly reduced (low level of evidence; 100% consensus) ejection fraction who have concomitant ID (defined as serum ferritin <100 ng/mL or serum ferritin 100–299 ng/mL with TSAT <20%).60) Practical advice on treatment and diagnosis of ID in patients with HF has previously been given61); further considerations are outlined in Figure 4.

Figure 4
Practical considerations for the diagnosis and treatment of iron deficiency in patients with heart failure.13, 31, 45, 61)
BNP = B-type natriuretic peptide; FCM = ferric carboxymaltose; FDI = ferric derisomaltose; Hb = haemoglobin; HF = heart failure; ID = iron deficiency; IV = intravenous; LVEF = left ventricular ejection fraction; NT-proBNP = N-terminal pro-B-type natriuretic peptide; TSAT = transferrin saturation.

*AFFIRM-AHF only enrolled patients during hospitalisation for an acute HF episode31); in addition to enrolling those patients, IRONMAN also enrolled non-hospitalised patients with elevated plasma NT-proBNP, elevated plasma BNP or with prior hospitalisation for HF within the last 6 months.45)

Note the 2021 European Society of Cardiology guidelines were updated prior to the availability of IRONMAN data, and so include a class IIa recommendation for use of IV FCM in patients with symptomatic HF, LVEF ≤50% and ID that have been recently hospitalised for HF based on the results of the AFFIRM-HF trial, but do not include FDI13); please refer to latest FCM and FDI Summary of Product Characteristics/Prescribing Information for approved indications and correct dosing.

In IRONMAN following initial treatment of ID (defined as serum ferritin <100 ng/mL and/or TSAT <20%), FDI was readministered if serum ferritin was <100 ng/mL, or ≤400 ng/mL when TSAT was <25% after 4 weeks and every 4 months with the aim of maintaining iron repletion between treatment visits45, 53); please note that IRONMAN uses a different definition of ID to the European Society of Cardiology and the Korean Society of Heart Failure Guidelines (serum ferritin values <100 ng/mL, or 100–299 ng/mL accompanied by TSAT <20%).

CONCLUSIONS AND FUTURE DIRECTIONS

ID is common in patients with acute or chronic HF, where it is associated with undesirable effects, such as exacerbating reductions in exercise capacity and QoL, and increased risk of HF hospitalisation. Data suggest a severe clinical phenotype in Asian patients with HF and ID, highlighting a need for early identification and treatment of ID in these patients. While the pathophysiology underlying ID in HF is unclear, evidence suggests that IV iron supplementation is beneficial for reducing hospitalisation rates and improving functional disease state, exercise capacity and QoL in patients with ID and HF. The upcoming FAIR-HF2 (NCT03036462) and HEART-FID (NCT03037931) clinical trials will provide further evidence regarding the effect of IV iron supplementation on the long-term CV mortality, exercise capacity and QoL in patients with HF and ID. Although analyses investigating the effect of IV iron in patients from Korea (or even from Asia in general) are limited, the effects of IV iron would not be expected to differ substantially in this population; nevertheless, clarifying studies may be of interest.

Notes

Funding:Medical writing support provided by Ebony Unogwu of AXON Communications (London, UK), funded by Vifor Pharma Management Ltd.

Conflict of Interest:Jankowska EA has received research grants and personal fees from Vifor Pharma (co-PI of the AFFIRM trial); and personal fees from Bayer, Novartis, Abbott, Boehringer Ingelheim, Pfizer, Servier, AstraZeneca, Berlin Chemie, Cardiac Dimensions, Fresenius, Respicardia, Zoll, Sanofi, Takeda, and Gedeon Richter. Ponikowski P has received research grants and personal fees from Vifor Pharma (PI of AFFIRM-AHF; participation in clinical trials); and personal fees from Amgen, Bayer, Novartis, Abbott Vascular, Boehringer Ingelheim, Pfizer, Servier, AstraZeneca, Berlin Chemie, Cibiem, BMS, Impulse Dynamics (participation in clinical trials).

Author Contributions:

  • Conceptualization: Jankowska EA, Ponikowski P.

  • Writing - original draft: Jankowska EA.

  • Writing - review & editing: Jankowska EA, Ponikowski P.

References

    1. Han X, Ding S, Lu J, Li Y. Global, regional, and national burdens of common micronutrient deficiencies from 1990 to 2019: a secondary trend analysis based on the Global Burden of Disease 2019 study. EClinicalMedicine 2022;44:101299
    1. Lippi G, Sanchis-Gomar F. Global epidemiology and future trends of heart failure. AME Med J 2020;5:15.
    1. Callender T, Woodward M, Roth G, et al. Heart failure care in low- and middle-income countries: a systematic review and meta-analysis. PLoS Med 2014;11:e1001699
    1. Park JJ, Lee CJ, Park SJ, et al. Heart failure statistics in Korea, 2020: a report from the Korean Society of Heart Failure. Int J Heart Fail 2021;3:224–236.
    1. Alcaide-Aldeano A, Garay A, Alcoberro L, et al. Iron deficiency: impact on functional capacity and quality of life in heart failure with preserved ejection fraction. J Clin Med 2020;9:1199.
    1. Bekfani T, Pellicori P, Morris D, et al. Iron deficiency in patients with heart failure with preserved ejection fraction and its association with reduced exercise capacity, muscle strength and quality of life. Clin Res Cardiol 2019;108:203–211.
    1. Jankowska EA, Rozentryt P, Witkowska A, et al. Iron deficiency: an ominous sign in patients with systolic chronic heart failure. Eur Heart J 2010;31:1872–1880.
    1. Jankowska EA, Kasztura M, Sokolski M, et al. Iron deficiency defined as depleted iron stores accompanied by unmet cellular iron requirements identifies patients at the highest risk of death after an episode of acute heart failure. Eur Heart J 2014;35:2468–2476.
    1. Van Aelst LN, Abraham M, Sadoune M, et al. Iron status and inflammatory biomarkers in patients with acutely decompensated heart failure: early in-hospital phase and 30-day follow-up. Eur J Heart Fail 2017;19:1075–1076.
    1. Yeo TJ, Yeo PS, Ching-Chiew Wong R, et al. Iron deficiency in a multi-ethnic Asian population with and without heart failure: prevalence, clinical correlates, functional significance and prognosis. Eur J Heart Fail 2014;16:1125–1132.
    1. Park JJ, Yoon M, Cho HW, et al. Iron deficiency in Korean patients with heart failure. J Korean Med Sci 2023;38:e177
    1. Parikh A, Natarajan S, Lipsitz SR, Katz SD. Iron deficiency in community-dwelling US adults with self-reported heart failure in the National Health and Nutrition Examination Survey III: prevalence and associations with anemia and inflammation. Circ Heart Fail 2011;4:599–606.
    1. McDonagh TA, Metra M, Adamo M, et al. 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J 2021;42:3599–3726.
    1. Rangel I, Gonçalves A, de Sousa C, et al. Iron deficiency status irrespective of anemia: a predictor of unfavorable outcome in chronic heart failure patients. Cardiology 2014;128:320–326.
    1. Andrews NC. Disorders of iron metabolism. N Engl J Med 1999;341:1986–1995.
    1. Goodnough LT, Nemeth E, Ganz T. Detection, evaluation, and management of iron-restricted erythropoiesis. Blood 2010;116:4754–4761.
    1. Jankowska EA, von Haehling S, Anker SD, Macdougall IC, Ponikowski P. Iron deficiency and heart failure: diagnostic dilemmas and therapeutic perspectives. Eur Heart J 2013;34:816–829.
    1. Brown DA, Perry JB, Allen ME, et al. Expert consensus document: mitochondrial function as a therapeutic target in heart failure. Nat Rev Cardiol 2017;14:238–250.
    1. Macdougall IC, Malyszko J, Hider RC, Bansal SS. Current status of the measurement of blood hepcidin levels in chronic kidney disease. Clin J Am Soc Nephrol 2010;5:1681–1689.
    1. Ueda N, Takasawa K. Impact of inflammation on ferritin, hepcidin and the management of iron deficiency anemia in chronic kidney disease. Nutrients 2018;10:1173.
    1. Wish JB, Aronoff GR, Bacon BR, et al. Positive iron balance in chronic kidney disease: how much is too much and how to tell? Am J Nephrol 2018;47:72–83.
    1. Jankowska EA, Malyszko J, Ardehali H, et al. Iron status in patients with chronic heart failure. Eur Heart J 2013;34:827–834.
    1. Weber CS, Beck-da-Silva L, Goldraich LA, Biolo A, Clausell N. Anemia in heart failure: association of hepcidin levels to iron deficiency in stable outpatients. Acta Haematol 2013;129:55–61.
    1. Anand IS, Gupta P. Anemia and iron deficiency in heart failure: current concepts and emerging therapies. Circulation 2018;138:80–98.
    1. Garcia-Casal MN, Pasricha SR, Martinez RX, Lopez-Perez L, Peña-Rosas JP. Serum or plasma ferritin concentration as an index of iron deficiency and overload. Cochrane Database Syst Rev 2021;5:CD011817
    1. Pasricha SR, Flecknoe-Brown SC, Allen KJ, et al. Diagnosis and management of iron deficiency anaemia: a clinical update. Med J Aust 2010;193:525–532.
    1. Wish JB. Assessing iron status: beyond serum ferritin and transferrin saturation. Clin J Am Soc Nephrol 2006;1 Suppl 1:S4–S8.
    1. Korean Society of Heart Failure. 2022 KSHF Guideline for the Management of Heart Failure. Seoul: Korean Society of Heart Failure; 2022.
    1. Anker SD, Comin Colet J, Filippatos G, et al. Ferric carboxymaltose in patients with heart failure and iron deficiency. N Engl J Med 2009;361:2436–2448.
    1. Ponikowski P, van Veldhuisen DJ, Comin-Colet J, et al. Beneficial effects of long-term intravenous iron therapy with ferric carboxymaltose in patients with symptomatic heart failure and iron deficiency. Eur Heart J 2015;36:657–668.
    1. Ponikowski P, Kirwan BA, Anker SD, et al. Ferric carboxymaltose for iron deficiency at discharge after acute heart failure: a multicentre, double-blind, randomised, controlled trial. Lancet 2020;396:1895–1904.
    1. Savarese G, Becher PM, Lund LH, Seferovic P, Rosano GM, Coats AJ. Global burden of heart failure: a comprehensive and updated review of epidemiology. Cardiovasc Res 2023;118:3272–3287.
    1. Zhang H, Zhabyeyev P, Wang S, Oudit GY. Role of iron metabolism in heart failure: from iron deficiency to iron overload. Biochim Biophys Acta Mol Basis Dis 2019;1865:1925–1937.
    1. Jankowska EA, Rozentryt P, Witkowska A, et al. Iron deficiency predicts impaired exercise capacity in patients with systolic chronic heart failure. J Card Fail 2011;17:899–906.
    1. Comín-Colet J, Enjuanes C, González G, et al. Iron deficiency is a key determinant of health-related quality of life in patients with chronic heart failure regardless of anaemia status. Eur J Heart Fail 2013;15:1164–1172.
    1. McEwan P, Ponikowski P, Davis JA, et al. Ferric carboxymaltose for the treatment of iron deficiency in heart failure: a multinational cost-effectiveness analysis utilising AFFIRM-AHF. Eur J Heart Fail 2021;23:1687–1697.
    1. Atherton JJ, Hayward CS, Wan Ahmad WA, et al. Patient characteristics from a regional multicenter database of acute decompensated heart failure in Asia Pacific (ADHERE International-Asia Pacific). J Card Fail 2012;18:82–88.
    1. Lam CS, Anand I, Zhang S, et al. Asian Sudden Cardiac Death in Heart Failure (ASIAN-HF) registry. Eur J Heart Fail 2013;15:928–936.
    1. Camaschella C. Iron deficiency. Blood 2019;133:30–39.
    1. Ambrosy AP, Lewis GD, Malhotra R, et al. Identifying responders to oral iron supplementation in heart failure with a reduced ejection fraction: a post-hoc analysis of the IRONOUT-HF trial. J Cardiovasc Med (Hagerstown) 2019;20:223–225.
    1. Lewis GD, Malhotra R, Hernandez AF, et al. Effect of oral iron repletion on exercise capacity in patients with heart failure with reduced ejection fraction and iron deficiency: the IRONOUT HF randomized clinical trial. JAMA 2017;317:1958–1966.
    1. Lyseng-Williamson KA, Keating GM. Ferric carboxymaltose: a review of its use in iron-deficiency anaemia. Drugs 2009;69:739–756.
    1. Pharmacosmos UK Limited. Ferric derisomaltose Pharmacosmos 100 mg/mL solution for injection/infusion - summary of product characteristics (SmPC) [Internet]. Leatherhead: Electronic Medicines Compendium; 2023 [cited 2023 June 16].
    1. Koduru P, Abraham BP. The role of ferric carboxymaltose in the treatment of iron deficiency anemia in patients with gastrointestinal disease. Therap Adv Gastroenterol 2016;9:76–85.
    1. Kalra PR, Cleland JG, Petrie MC, et al. Intravenous ferric derisomaltose in patients with heart failure and iron deficiency in the UK (IRONMAN): an investigator-initiated, prospective, randomised, open-label, blinded-endpoint trial. Lancet 2022;400:2199–2209.
    1. Macdougall IC. Evolution of iv iron compounds over the last century. J Ren Care 2009;35 Suppl 2:8–13.
    1. Beck-da-Silva L, Piardi D, Soder S, et al. IRON-HF study: a randomized trial to assess the effects of iron in heart failure patients with anemia. Int J Cardiol 2013;168:3439–3442.
    1. Okonko DO, Grzeslo A, Witkowski T, et al. Effect of intravenous iron sucrose on exercise tolerance in anemic and nonanemic patients with symptomatic chronic heart failure and iron deficiency FERRIC-HF: a randomized, controlled, observer-blinded trial. J Am Coll Cardiol 2008;51:103–112.
    1. Comin-Colet J, Lainscak M, Dickstein K, et al. The effect of intravenous ferric carboxymaltose on health-related quality of life in patients with chronic heart failure and iron deficiency: a subanalysis of the FAIR-HF study. Eur Heart J 2013;34:30–38.
    1. Anker SD, Ponikowski P, Khan MS, et al. Responder analysis for improvement in 6-min walk test with ferric carboxymaltose in patients with heart failure with reduced ejection fraction and iron deficiency. Eur J Heart Fail 2022;24:833–842.
    1. Butler J, Khan MS, Friede T, et al. Health status improvement with ferric carboxymaltose in heart failure with reduced ejection fraction and iron deficiency. Eur J Heart Fail 2022;24:821–832.
    1. Jankowska EA, Kirwan BA, Kosiborod M, et al. The effect of intravenous ferric carboxymaltose on health-related quality of life in iron-deficient patients with acute heart failure: the results of the AFFIRM-AHF study. Eur Heart J 2021;42:3011–3020.
    1. Kalra PR, Cleland JG, Petrie MC, et al. Rationale and design of a randomised trial of intravenous iron in patients with heart failure. Heart 2022;108:1979–1985.
    1. Lim EA, Sohn HS, Lee H, Choi SE. Cost-utility of ferric carboxymaltose (Ferinject®) for iron-deficiency anemia patients with chronic heart failure in South Korea. Cost Eff Resour Alloc 2014;12:19.
    1. Yeo TJ, Yeo PS, Hadi FA, et al. Single-dose intravenous iron in Southeast Asian heart failure patients: a pilot randomized placebo-controlled study (PRACTICE-ASIA-HF). ESC Heart Fail 2018;5:344–353.
    1. Jankowska EA, Tkaczyszyn M, Suchocki T, et al. Effects of intravenous iron therapy in iron-deficient patients with systolic heart failure: a meta-analysis of randomized controlled trials. Eur J Heart Fail 2016;18:786–795.
    1. Toblli JE, Lombraña A, Duarte P, Di Gennaro F. Intravenous iron reduces NT-pro-brain natriuretic peptide in anemic patients with chronic heart failure and renal insufficiency. J Am Coll Cardiol 2007;50:1657–1665.
    1. Anker SD, Kirwan BA, van Veldhuisen DJ, et al. Effects of ferric carboxymaltose on hospitalisations and mortality rates in iron-deficient heart failure patients: an individual patient data meta-analysis. Eur J Heart Fail 2018;20:125–133.
    1. Graham FJ, Pellicori P, Ford I, Petrie MC, Kalra PR, Cleland JG. Intravenous iron for heart failure with evidence of iron deficiency: a meta-analysis of randomised trials. Clin Res Cardiol 2021;110:1299–1307.
    1. Sim D, Lin W, Sindone A, et al. Asian Pacific Society of Cardiology consensus statements on the diagnosis and management of chronic heart failure. J Asia Pac Soc Cardiol 2023;2:e10
    1. Sindone A, Doehner W, Manito N, et al. Practical guidance for diagnosing and treating iron deficiency in patients with heart failure: why, who and how? J Clin Med 2022;11:2976.

Metrics
Share
Figures

1 / 4

Tables

1 / 1

Funding Information
PERMALINK