Article

CONFIRM-HF – Targeting for Improvement in Exercise Capacity in Heart Failure

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.
Information image
Average (ratings)
No ratings
Your rating

Disclosure:Piotr Ponikowski has received honoraria from Vifor Pharma as a member of the FAIR-HF and CONFIRM-HF steering committees; consultancy and speakers bureau from Vifor Pharma and Amgen Inc; and a research grant from Vifor Pharma.

Received:

Accepted:

Published online:

Support:Vifor Pharma funded a writer to attend the symposium and develop this manuscript accordingly.

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.

ID is a frequent co-morbidity in stable HF and in patients admitted to hospital due to HF worsening.3,6,21,22 Its association with impaired exercise capacity, poor QoL and increased mortality, irrespective of anaemia,3–5 make it an attractive therapeutic target – a hypothesis that has recently been tested in clinical studies.22 Several options are available for the correction of ID. Blood transfusion is generally not recommended since it is associated with high mortality and a lengthy stay in hospital;23 its use should be reserved for life-threatening emergency situations. Erythropoiesis-stimulating agents (ESA) are used mainly to correct anaemia. In the Reduction of Events with Darbepoetin alfa in Heart Failure (RED-HF) study, a randomised, double-blind trial on anaemic patients with chronic HF (n=2,278), correcting anaemia with darbepoetin alpha did not lead to improvement in survival nor clinically meaningful change in QoL.24 Furthermore, patients treated with darbepoetin alpha had an increased risk for thromboembolic adverse effects.24 One potential explanation of the neutral treatment effects on the outcomes could be due to the fact that ESA therapy can further exacerbate ID by stimulating the production of red blood cells, which requires a large amount of iron. Therefore, there could be a subpopulation of patients included in the RED-HF study with underlying untreated ID based on the current definition of ID in the ESC Guidelines for HF. Post hoc sub-analysis investigating the association between ID and the outcomes in the RED-HF trial is undergoing.

Another option for correcting ID is iron therapy, which may be administered by oral or i.v. routes. There is no evidence for the clinical benefits of oral iron supplementation – studies comparing oral iron with ESA in patients with HF and anaemia found no clinically meaningful benefits associated with such combination.25–29 A recent pilot study (n=18) suggested that i.v. and not oral iron improves exercise capacity in HF patients.30 Early clinical studies have also demonstrated the efficacy of i.v. therapy but were either single-arm, open-label studies with a short-term follow-up or small sample size.31–34 A larger, randomised, double-blind, placebo-controlled trial was therefore needed. Therefore, the FAIR-HF (n=459) trial was performed and showed that treatment of HF patients with i.v FCM in patients with chronic HF and ID, with or without anaemia, improved symptoms, exercise capacity and Qol at six months, including significant improvements in self-reported patient global assessment (PGA), NYHA functional class, six-minute walk test (6MWT), Kansas City Cardiomyopathy Questionnaire (KCCQ) overall score and EQ-5D visual analogue scale (VAS) score.22 Improvements were seen from week 4 onwards irrespective of whether or not the patients were anaemic.

In order to broaden the evidence in support to treat ID with i.v. iron in HF, it was necessary to replicate these findings in a further study, and also to evaluate different, more robust and objective endpoints, including safety endpoints as well as a longer follow-up. In addition, the FAIR-HF study employed repeated 200 mg doses, therefore an evaluation of higher single-dose (up to 1,000 mg) was needed. To address these questions, the CONFIRM-HF clinical trial was designed.

CONFIRM-HF was a multicentre, randomised (1:1) double-blind, placebo-controlled trial of stable, ambulatory HF patients.35 The main inclusion criteria were:

  • NYHA class II/II with left ventricle ejection fraction (LVEF) ≤45 %;
  • Brain natriuretic peptide (BNP) >100 pg/mL or the prohormone NT-proBNP >400 pg/mL;
  • ID, defined as serum ferritin <100 ng/mL or 100–300 ng/mL if TSAT <20 %; and
  • haemoglobin (Hb) <15 g/dL and no lower Hb cut-off level.

The CONFIRM-HF Study – Change in Six-minute Walking Test Distance at 24 Weeks

Article image

The CONFIRM-HF Study – First Hospitalisation Due to Worsening Heart Failure

Article image

Patients who needed a blood transfusion were excluded from the study. The methodology involved blinded and unblinded personnel and the usage of curtains and black syringes for injections, to mask the brownish colour of FCM. Treatment involved a correction phase (given as 1–2 i.v. injections of 500–1,000 iron as single dose treatment (500 mg) continued (at weeks 12, 24 and 36) if ID was not corrected. The primary endpoint was the change in 6MWT distance from baseline to Week 24. Key secondary endpoints included the 6MWT distance at Week 6, 12, 36 and 52; PGA score, NYHA class, KCCQ, EQ-5D and fatigue score at Week 6, 12, 24, 36 and 52. Outcome-related secondary endpoints included hospitalisation rate (all hospitalisation, for any cardiovascular [CV] reason, due to worsening HF); time to first hospitalisation (all hospitalisation, for any CV reason and due to worsening HF); and time to death (any death, death for any CV reason and due to worsening HF).35 At Week 24, there was an increase in 6MWT distance by 18 ± 8 m in the FCM group, whereas in the placebo group 6MWT distance decreased by 16 ± 8 m (both least squares mean + standard error [SE]). It resulted in a significant increase in 6MWT distance at Week 24 in FCM by 33 ± 11 m (least squares mean ± SE) compared with placebo (p=0.002, see Figure 4), a treatment effect that had only previously been seen for cardiac resynchronisation therapy. In all subgroups examined, including those patients with and without anaemia, the treatment effect was preserved. The improvement in exercise capacity was striking in that it was reported not only at 24 weeks, but also at 36 and 52 weeks, with a sustained improvement in fatigue score also observed. Throughout the study, improvements in other secondary endpoints in patients treated with FCM were detected with statistical significance observed from Week 24 onwards. Treatment with FCM was associated with a significant reduction in the risk of hospitalisations for worsening HF (hazard ratio [95 % confidence interval]: 0.39 [0.19–0.82], p=0.009; see Figure 5). The number of deaths (FCM: 12, placebo: 14 deaths) and the incidence of adverse events were comparable between both groups.35

In conclusion, the CONFIRM-HF trial demonstrated that in symptomatic patients with chronic HF and ID treatment with i.v. FCM over one year resulted in sustainable improvements in exercise capacity, symptoms and QoL and may be associated with a reduced risk of hospitalisations due to worsening HF. Further studies are planned and ongoing, including a meta-analysis, the EFfect of Ferric carboxymaltose on Exercise Capacity in PaTients with iron deficiency and chronic Heart Failure (EFFECT-HF), iron in Congestive Heart Failure (iCHF)36 and the Ferric Carboxymaltose Assessment in Patients With IRon Deficiency and Chronic Heart Failure with preserved ejection fraction (FAIR-HFpEF) studies.37 Based on current data, a suggested treatment algorithm has been proposed (see Figure 6).

Suggested Treatment Algorithm for Iron Deficiency in Heart Failure, Based on Evidence from Clinical Trials

Article image

References

  1. Lesman-Leegte I, Jaarsma T, Coyne JC, et al., Quality of life and depressive symptoms in the elderly: a comparison between patients with heart failure and age- and gender-matched community controls, J Card Fail, 2009;15:17–23.
    Crossref | Pubmed
  2. McMurray JJ, Adamopoulos S, Anker SD, et al., ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC, Eur Heart J, 2012;33:1787–847.
    Crossref | Pubmed
  3. 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
  4. 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
  5. Comin-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;15:1164–72.
    Crossref | Pubmed
  6. Enjuanes C, Klip IT, Bruguera J, et al., Iron deficiency and health-related quality of life in chronic heart failure: results from a multicenter European study, Int J Cardiol, 2014;174:268–75.
    Crossref | Pubmed
  7. McMurray JJ, Adamopoulos S, Anker SD, et al., ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC, Eur J Heart Fail, 2012;14:803–69.
    Crossref | Pubmed
  8. 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;16:1125–32.
    Crossref | Pubmed
  9. Zijp IM, Korver O, Tijburg LB, Effect of tea and other dietary factors on iron absorption, Crit Rev Food Sci Nutr, 2000;40:371–98.
    Crossref | Pubmed
  10. Finberg KE, Heeney MM, Campagna DR, et al., Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA), Nat Genet, 2008;40:569–71.
    Crossref | Pubmed
  11. An P, Wu Q, Wang H, et al., TMPRSS6, but not TF, TFR2 or BMP2 variants are associated with increased risk of iron-deficiency anemia, Hum Mol Genet, 2012;21:2124–31.
    Crossref | Pubmed
  12. Andrews NC, Disorders of iron metabolism, N Engl J Med, 1999;341:1986–95.
    Crossref | Pubmed
  13. 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
  14. Ganz T, Hepcidin and its role in regulating systemic iron metabolism, Hematology Am Soc Hematol Educ Program, 2006;29–35, 507.
    Crossref | Pubmed
  15. Zhang AS, Enns CA, Molecular mechanisms of normal iron homeostasis, Hematology Am Soc Hematol Educ Program, 2009;207–14.
    Crossref | Pubmed
  16. Oexle H, Gnaiger E, Weiss G, Iron-dependent changes in cellular energy metabolism: influence on citric acid cycle and oxidative phosphorylation, Biochim Biophys Acta, 1999;1413:99–107.
    Crossref | Pubmed
  17. Haas JD, Brownlie T 4th, Iron deficiency and reduced work capacity: a critical review of the research to determine a causal relationship, J Nutr, 2001;131:676S–88S; discussion 688S–90S.
    Pubmed
  18. Dallman PR, Iron deficiency: does it matter?, J Intern Med, 1989;226:367–72.
    Crossref | Pubmed
  19. Yeo TJ, Yeo PS, Sim DKL, et al., Functional iron deficiency in heart failure with preserved versus reduced ejection fraction, J Am Coll Cardiol, 2014;63(12 S):A778.
    Crossref
  20. Krum H, Jelinek MV, Stewart S, et al., 2011 update to National Heart Foundation of Australia and Cardiac Society of Australia and New Zealand Guidelines for the prevention, detection and management of chronic heart failure in Australia, 2006, Med J Aust, 2011;194:405–9.
    Pubmed
  21. 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
  22. 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
  23. Kao DP, Kreso E, Fonarow GC, Krantz MJ, Characteristics and outcomes among heart failure patients with anemia and renal insufficiency with and without blood transfusions (public discharge data from California 2000-2006), Am J Cardiol, 2011;107:69–73.
    Crossref | Pubmed
  24. Swedberg K, Young JB, Anand IS, et al., Treatment of anemia with darbepoetin alfa in systolic heart failure, N Engl J Med, 2013;368:1210–9.
    Crossref | Pubmed
  25. Ghali JK, Anand IS, Abraham WT, et al., Randomized double-blind trial of darbepoetin alfa in patients with symptomatic heart failure and anemia, Circulation, 2008;117:526–35.
    Crossref | Pubmed
  26. Parissis JT, Kourea K, Panou F, et al., Effects of darbepoetin alpha on right and left ventricular systolic and diastolic function in anemic patients with chronic heart failure secondary to ischemic or idiopathic dilated cardiomyopathy, Am Heart J, 2008;155:751 e1–7.
    Crossref | Pubmed
  27. van Veldhuisen DJ, Dickstein K, Cohen-Solal A, et al., Randomized, double-blind, placebo-controlled study to evaluate the effect of two dosing regimens of darbepoetin alfa in patients with heart failure and anaemia, Eur Heart J, 2007;28:2208–16.
    Crossref | Pubmed
  28. Kourea K, Parissis JT, Farmakis D, et al., Effects of darbepoetin-alpha on quality of life and emotional stress in anemic patients with chronic heart failure, Eur J Cardiovasc Prev Rehabil, 2008;15:365–9.
    Crossref | Pubmed
  29. Palazzuoli A, Silverberg DS, Iovine F, et al., Effects of beta-erythropoietin treatment on left ventricular remodeling, systolic function, and B-type natriuretic peptide levels in patients with the cardiorenal anemia syndrome, Am Heart J, 2007;154:645 e9–15.
    Crossref | Pubmed
  30. 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;168:3439–42.
    Crossref | Pubmed
  31. Bolger AP, Bartlett FR, Penston HS, et al., Intravenous iron alone for the treatment of anemia in patients with chronic heart failure, J Am Coll Cardiol, 2006;48:1225–7.
    Crossref | Pubmed
  32. 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;50:1657–65.
    Crossref | Pubmed
  33. 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;51:103–12.
    Crossref | Pubmed
  34. Usmanov RI, Zueva EB, Silverberg DS, Shaked M, Intravenous iron without erythropoietin for the treatment of iron deficiency anemia in patients with moderate to severe congestive heart failure and chronic kidney insufficiency, J Nephrol, 2008;21:236–42.
    Pubmed
  35. 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, 2014 [Epub ahead of print].
    Crossref | Pubmed
  36. Iron in Congestive Heart Failure (iCHF). Available at: http://clinicaltrials.gov/ct2/show/NCT01837082?term=iCHF&rank=1 (accessed 17 September 2014).
  37. EFfect of Ferric carboxymaltose on Exercise Capacity in PaTients with iron deficiency and chronic Heart Failure (EFFECT-HF). Available at: http://clinicaltrials.gov/show/NCT01394562 (accessed 3 September 2014).