Article

Understanding Iron Deficiency in Heart Failure: Clinical Significance and Management

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

Iron deficiency (ID) has been increasingly recognized as an important co-morbidity associated with heart failure (HF). ID significantly impairs exercise tolerance and is an independent predictor of poor outcomes in people with HF irrespective of their anemic status. Diagnosis of ID in people with HF is often missed and therefore routine screening for ID is necessary for these patients. IV iron repletion has been recommended in HF treatment guidelines to improve symptoms and exercise capacity. People with ID and HF who are treated with IV iron have an improved quality of life, better 6-minute walk test results and New York Heart Association functional class. The effect of iron therapy on re-hospitalization and mortality rates in people with HF remains unclear. Large-dose oral iron treatment has been found to be ineffective in improving symptoms in people with HF. This review summarizes the current knowledge on prevalence, clinical relevance, and the molecular mechanism of ID in patients with chronic HF and the available evidence for the use of parenteral iron therapy.

Disclosure:The author has no conflict of interest to declare.

Received:

Accepted:

Correspondence Details:Rajalakshmi Santhanakrishnan, 235A Biological Sciences, Wright State University, Dayton, OH 45431, USA. E: santhanakrishnan.2@wright.edu

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Heart failure (HF) is a major health problem and about 6 million adults are affected in the US alone, at a cost of approximately $20 billion per year.1 Despite the availability of new treatment strategies, the incidence, number of hospitalizations and mortality associated with HF remains a big health burden.1 In addition to increasing age, the factors that contribute to poor prognosis for people with HF are its associated co-morbidities. Iron deficiency (ID) has been increasingly recognized as an important co-morbidity that contributes to increased incidence, re-hospitalization and poor survival in people with HF.2 Anemia is the ultimate consequence of ID, but the conditions are distinct clinical scenarios. Identification of ID in people with chronic diseases such as HF is important as iron repletion has shown to improve patient’s symptoms irrespective of anemic status.

Mechanism of Iron Deficiency in Heart Failure

Iron is an essential micronutrient in all types of cells and is particularly important in energy-demanding cells such as cardiomyocytes.3 Iron acts as a co-factor for several enzymes involved in oxidative phosphorylation and plays a key role in oxygen transport through erythropoiesis. The etiology of ID in HF is not clearly understood; it could possibly be due to multiple factors including poor iron absorption due to edema in the gastrointestinal tract, low bioavailability of iron, and a chronic inflammatory state.4 At the molecular level, one possible mechanism of ID is related to hepcidin, an iron regulatory hormone, whose synthesis is stimulated when iron stores or levels of the cytokine interlukin-6 (IL-6) are elevated. In chronic inflammatory conditions, hepcidin levels are increased due to an increase in IL-6 and/or fluctuating iron levels. Elevated hepcidin results in removal of ferroportin – a protein that increases iron efflux into the bloodstream – from the duodenal surfaces.5 However, in HF, a reverse mechanism exists: initially hepcidin level increases but as HF progresses, hepcidin is downregulated which maintains ferroportin levels thereby increasing iron efflux.6 The reason for this is still not clearly understood. Importantly, lower hepcidin levels have been associated with increased 3-year mortality rate in people with acute HF.7

Another mechanism underlying ID in people with HF is thought to be liver congestion that leads to increased hemosiderin-laden macrophages.8 Cardiomyocytes have a high-energy demand and are therefore susceptible to ID. In patients referred for cardiac transplantation, myocardial iron stores have been found to be lower compared with patients without HF.9 HF patients also showed reduced myocardial oxygen respiration and reduction in mitochondrial respiratory enzymes.10 In experimental mice models, the left ventricular cardiomyocytes with depleted iron-regulatory protein (IRP), which maintains intracellular iron availability, showed reduced mitochondrial complex I activity and the mice were unable to increase ventricular systolic function in response to dobutamine stress.11 Despite there being different possibilities for the mechanism underlying ID in HF disease, its pathophysiology is unclear.

Prevalence of Iron Deficiency in Heart Failure

The prevalence of ID has been widely studied in patients with chronic HF with reduced ejection fraction (HFrEF) and ranges between 36 and 69 % have been found among different ethnic groups with the Portuguese having the lowest and British having the highest prevalence rates.12

In patients presenting with acute decompensated HF (ADHF), the prevalence of ID is much higher than those with chronic HF.7 A gender difference in the prevalence of ID was shown in a French study of 832 people with ADHF; 66 % of men and 75 % of women had ID.13 Independent correlates of ID in HF include increased age, higher New York Heart Association (NYHA) functional class, being a woman, elevated N-terminal pro-B-type natriuretic peptide (NT-proBNP), and high sensitivity C-reactive protein.2 The prevalence of ID in people with HFpEF is still unknown.

Consequences of Iron Deficiency

Heart Failure with Reduced Ejection Fraction

ID greatly decreases the quality of life irrespective of the presence of anemia in patients with HFrEF. In one study, patients with concurrent ID and HFrEF had lower peak oxygen consumption (peak VO2) and increased ventilator response to exercise (VE-VCO2 slope) compared with those without concurrent ID with both reflecting poor exercise capacity.14 Quality of life has been shown to be significantly affected in HF patients irrespective of diagnostic criteria using either the European Quality of Life-5D, Kansas City Cardiomyopathy, or the Minnesota Living with Heart Failure questionnaires.15

In one study, of 1,506 HF patients, short-term 6-month follow-up showed a higher risk of death in those with HF and ID compared with patients with HF and no ID (8.7 % versus 3.6 % respectively, p<0.001).16 Similarly, in long-term follow-up over approximately 2.5 years, those with ID had a higher likelihood of dying than those without ID.16 The association with mortality was independent of anemic status.

In another study involving 157 people with HF, participants with non-anemic ID had twice the risk of death than anemic patients on iron therapy.17 In a study that used serum-soluble transferrin receptor (sTfR) and hepcidin levels to define ID in people with ADHF, there was a 5 % in-hospital mortality and a strikingly higher risk of death (95 % CI [2.97–14.62], p<0.001) within 12 months of discharge.12

Heart Failure with Preserved Ejection Fraction

The definitive role of ID in patients with HF with preserved ejection faction (HFpEF) is poorly understood. In a small study involving 26 HFpEF subjects, neither cardiac function (both systolic and diastolic) nor reduced exercise capacity was dependent on ID.18 In another study involving 40 HFpEF patients, ID was a predictor of decreased exercise tolerance independent of ventricular diastolic function, renal function, hemoglobin and NT-proBNP.19 There was a significant correlation of both transferrin and ferritin levels with peak VO2 in these subjects. The difference in results between the two studies might be because the latter study involved subjects with more advanced disease (higher NT-proBNP, higher NYHA class, previous history of ADHF and relatively lower peak VO2). More studies are required to identify the significance of ID in people with HFpEF.

Diagnosis of Iron Deficiency in Heart Failure

Diagnosis of ID in HF is complicated by the fact that the symptoms of fatigue and exercise intolerance associated with ID overlap with the symptoms of HF. ID is defined as absolute and functional depending on the serum ferritin concentration (a marker of iron stores) and saturation level of transferrin – the iron transport protein. In the general population, a serum ferritin cut-off value of 30 µg/l is used for the diagnosis of ID.20 Ferritin is an acute-phase reactant, therefore a higher cut-off serum level is used for diagnosis of ID in people with chronic inflammatory state. Absolute ID is defined as reduced iron stores despite normal iron homeostasis diagnosed as serum ferritin level of <30 µg/l (Table 1). Functional ID is defined as an inability to meet the body’s iron demand despite having normal iron stores. It is measured by a combination of serum ferritin (100–300 µg/l) and transferrin saturation (<20 %) levels.3 The gold standard assay to diagnose ID is the bone marrow Prussian blue stain to identify absence of iron granules. It is an invasive procedure and is not routinely used.

Diagnosis of Iron Deficiency in Heart Failure

Article image

Guidelines for the Treatment of Iron Deficiency in Heart Failure

Article image

Iron Therapy in Heart Failure

Parenteral Iron

Having proven the significant correlation of ID in HF patients, several clinical trials of iron therapy have been conducted. The current treatment guidelines for HF with ID is listed in Table 2 and is based on two large trials: Ferinject® Assessment in Patients With IRon Deficiency and Chronic Heart Failure (FAIR-HF) and A Study to Compare the Use of Ferric Carboxymaltose With Placebo in Patients With Chronic Heart Failure and Iron Deficiency (CONFIRM-HF).21,22 Both trials tested intravenous ferric carboxymaltose (FCM) in patients with ambulatory chronic systolic HF with ID in NYHA classes II and III. The FAIR-HF trial included 459 HFrEF patients (EF <45 %) who completed a self-reported Patient Global Assessment, which improved in 50 % (n=147) of patients in the FCM group compared with 28 % (n=41) of the placebo group, along with improvement in NYHA functional class, 6-minute walk test and quality-of-life assessments. The positive changes were present in patients with ID both with and without anemia. CONFIRM-HF included 304 HFrEF patients and after 6 months of iron treatment, the primary endpoint of a 6-minute walk test significantly improved in the FCM group compared with the placebo group. All secondary endpoints including NYHA class, quality of life, fatigue score and time for first rehospitalization improved significantly in the FCM group. Both trials did not show any adverse side-effects with IV iron therapy. Although individual clinical trials were not tested for major clinical events, a meta-analysis including FAIR-HF, CONFIRM-HF, EFfect of Ferric Carboxymaltose on exercIse CApacity and Cardiac Function in Patients With Iron deficiencY and Chronic Heart Failure (EFFICACY-HF) and FER-CARS-01 assessed the effect of iron therapy in cardiovascular hospitalization, and mortality in patients with HF and ID.23 Treatment with IV FCM had lower rates of combined HF hospitalizations and cardiovascular mortality (rate ratio 0.53, 95 % CI [0.33–0.86], p=0.011). These findings from the meta-analysis need to be validated in randomized clinical trials.

Oral Iron

The Iron Repletion Effects on Oxygen Uptake in Heart Failure (IRONOUT HF) study is a large-scale randomized clinical trial that tested the role of high-dose oral iron therapy in 225 patients with HFrEF and ID.24 The primary endpoint was change in peak VO2 at 16 weeks and secondary endpoints included change in NT-proBNP, 6-minute walk test and quality of life. In contrast to results from trials that used IV iron, the IRONOUT HF trial failed to show improvements in primary and secondary endpoints.

Conclusion

ID is more common in patients with HF and is often overlooked because of overlapping clinical symptoms. ID in HF patients is associated with reduced exercise tolerance, poor quality of life and increased rehospitalizations and mortality. Routine screening for ID in patients with HF and treatment with iron is recommended by HF treatment guidelines.1 Parenteral iron improves patient symptoms, quality of life and exercise capacity after iron therapy.26 Despite improvements in HF symptoms, major clinical outcomes were not tested in clinical trials. Further, all trials included only patients with HFrEF and the role of ID in patients with HFpEF is not clearly understood. More randomized clinical trials that might answer the open questions on the effect of iron repletion on long-term symptoms, effects on rehospitalization and survival in patients with concurrent ID and HF are currently underway.

References

  1. Yancy CW, Jessup M, Bozkurt B, et al. 2017 ACC/AHA/HFSA Focused Update of the 2013 ACCF/AHA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. J Card Fail 2017;23:628-651.
    Crossref | PubMed
  2. Yeo TJ, Yeo PS, Ching-Chiew WR, 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;16:1125–32.
    Crossref | PubMed
  3. Cappellini MD, Comin-Colet J, de Francisco A, et al. Iron deficiency across chronic inflammatory conditions: International expert opinion on definition, diagnosis, and management. Am J Hematol 2017;92:1068–78.
    Crossref | PubMed
  4. Jankowska EA, Von Haehling S, Anker SD, et al. Iron deficiency and heart failure: diagnostic dilemmas and therapeutic perspectives. Eur Heart J 2013;34:816–29.
    Crossref | PubMed
  5. Ganz T, Nemeth E. The hepcidin-ferroportin system as a therapeutic target in anemias and iron overload disorders. Hematology Am Soc Hematol Educ Program 2011;2011:538–42.
    Crossref | PubMed
  6. Jankowska EA, Malyszko J, Ardehali H, et al. Iron status in patients with chronic heart failure. Eur Heart J 2013;34:827–34.
    Crossref | PubMed
  7. 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;35:2468–76.
    Crossref | PubMed
  8. Suzuki T, Hanawa H, Jiao S, et al. Inappropriate expression of hepcidin by liver congestion contributes to anemia and relative iron deficiency. J Card Fail 2014;20:268–77.
    Crossref | PubMed
  9. Leszek P, Sochanowicz B, Szperl M, et al. Myocardial iron homeostasis in advanced chronic heart failure patients. Int J Cardiol 2012;159:47–52.
    Crossref | PubMed
  10. Melenovsky V, Petrak J, Mracek T, et al. Myocardial iron content and mitochondrial function in human heart failure: a direct tissue analysis. Eur J Heart Fail 2017;19:522–30.
    Crossref | PubMed
  11. Haddad S, Wang Y, Galy B, et al. Iron-regulatory proteins secure iron availability in cardiomyocytes to prevent heart failure. Eur Heart J 2017;38:362–72.
    Crossref | PubMed
  12. Fitzsimons S, Doughty RN. Iron deficiency in patients with heart failure. Eur Hear J Cardiovasc Pharmacother 2015;1:58-64.
    Crossref | PubMed
  13. Cohen-Solal A, Damy T, Terbah M, et al. High prevalence of iron deficiency in patients with acute decompensated heart failure. Eur J Heart Fail 2014;16:984–91.
    Crossref | PubMed
  14. 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.
    Crossref | PubMed
  15. 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;18:786–95.
    Crossref | PubMed
  16. Klip IT, Comin-Colet J, Voors AA, et al. Iron deficiency in chronic heart failure: an international pooled analysis. Am Heart J 2013;165:575–82.e3.
    Crossref | PubMed
  17. Okonko DO, Mandal AKJ, Missouris CG, Poole-Wilson PA. Disordered iron homeostasis in chronic heart failure: prevalence, predictors, and relation to anemia, exercise capacity, and survival. J Am Coll Cardiol 2011;58:1241–51.
    Crossref | PubMed
  18. Kasner M, Aleksandrov AS, Westermann D, et al. Functional iron deficiency and diastolic function in heart failure with preserved ejection fraction. Int J Cardiol 2013;168:4652–7.
    Crossref | PubMed
  19. Núñez J, Domínguez E, Ramón JM, et al. Iron deficiency and functional capacity in patients with advanced heart failure with preserved ejection fraction. Int J Cardiol 2016;207:365–7.
    Crossref | PubMed
  20. Bermejo F, García-López S. A guide to diagnosis of iron deficiency and iron deficiency anemia in digestive diseases. World J Gastroenterol 2009;15:4638-43.
    Crossref | PubMed
  21. Anker SD, Comin Colet J, Filippatos G, et al. Ferric carboxymaltose in patients with heart failure and iron deficiency. N Engl J Med 2009;361:2436–48.
    Crossref | PubMed
  22. 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;36:657–68.
    Crossref | PubMed
  23. 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 2017;20:125–33.
    Crossref | PubMed
  24. 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. JAMA 2017;317:1958–66.
    Crossref | PubMed
  25. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 2016;37:2129–200.
    Crossref | PubMed