A prospective, multi-center, randomized comparison of iron isomaltoside 1000 versus iron sucrose in patients with iron deficiency anemia; the FERWON-IDA trial

Michael Auerbach, David Henry, Richard J Derman, Maureen M Achebe, Lars L Thomsen, John Glaspy, Michael Auerbach, David Henry, Richard J Derman, Maureen M Achebe, Lars L Thomsen, John Glaspy

Abstract

Iron deficiency anemia (IDA) is prevalent, and intravenous iron, especially if given in a single dose, may result in better adherence compared with oral iron. The present trial (FERWON-IDA) is part of the FERWON program with iron isomaltoside 1000/ferric derisomaltose (IIM), evaluating safety and efficacy of high dose IIM in IDA patients of mixed etiologies. This was a randomized, open-label, comparative, multi-center trial conducted in the USA. The IDA patients were randomized 2:1 to a single dose of 1000 mg IIM, or iron sucrose (IS) administered as 200 mg intravenous injections, up to five times. The co-primary endpoints were adjudicated serious or severe hypersensitivity reactions, and change in hemoglobin from baseline to week eight. A total of 1512 patients were enrolled. The frequency of patients with serious or severe hypersensitivity reactions was 0.3% (95% confidence interval: 0.06;0.88) vs 0.4% (0.05;1.45) in the IIM and IS group, respectively. The co-primary safety objective was met, and no risk difference was observed between groups. The co-primary efficacy endpoint of non-inferiority in hemoglobin change was met, and IIM led to a significantly more rapid hematological response in the first two weeks. The frequency of cardiovascular events was 0.8% and 1.2% in the IIM and IS group, respectively (P = .570). The frequency of hypophosphatemia was low in both groups. Iron isomaltoside administered as 1000 mg resulted in a more rapid and more pronounced hematological response, compared with IS, which required multiple visits. The safety profile was similar with a low frequency of hypersensitivity reactions and cardiovascular events.

Conflict of interest statement

Michael Auerbach receives research funding for data management from AMAG Pharmaceuticals.

David Henry has no conflict of interest.

Richard Derman has been a research and science consultant with Pharmacosmos and AMAG.

Lars L. Thomsen is employed by Pharmacosmos A/S.

Maureen M. Achebe has been on scientific advisory boards for Pharmacosmos and AMAG pharmaceuticals, and a consultant for Luitpold.

John Glaspy has been an advisor to AMAG Pharmaceuticals.

This work was funded by Pharmacosmos A/S and the investigators/institutions received a fee per patient.

© 2019 The Authors. American Journal of Hematology published by Wiley Periodicals, Inc.

Figures

Figure 1
Figure 1
Patient disposition. IIM, iron isomaltoside 1000/ferric derisomaltose; IS, iron sucrose
Figure 2
Figure 2
Hemoglobin, s‐ferritin, and transferrin saturation over time by treatment group (intention to treat analysis set). Estimates (mean and SE) from a mixed model with repeated measures with strata, treatment and time as factors, treatment*time and baseline value*time interactions and baseline value as covariate. IMM, iron isomaltoside 1000/ferric derisomaltose; IS, iron sucrose. *P < .05, **P < .001, ***P < .001

References

    1. Dignass AU, Gasche C, Bettenworth D, et al. European consensus on the diagnosis and management of iron deficiency and anaemia in inflammatory bowel diseases. J Crohns Colitis. 2015;9:211‐222.
    1. KDIGO Clinical Practice Guideline for Anemia in Chronic Kidney Disease. 2012. . Accessed May 22, 2019.
    1. Gasche C, Berstad A, Befrits R, et al. Guidelines on the diagnosis and management of iron deficiency and anemia in inflammatory bowel diseases. Inflamm Bowel Dis. 2007;13:1545‐1553.
    1. Macdougall IC, Bircher AJ, Eckardt KU, et al. Iron management in chronic kidney disease: conclusions from a “kidney disease: improving global outcomes” (KDIGO) controversies conference. Kidney Int. 2016;89:28‐39.
    1. Macdougall IC, Vernon K. Complement activation‐related pseudo‐allergy: a fresh look at hypersensitivity reactions to intravenous iron. Am J Nephrol. 2017;45:60‐62.
    1. Hempel JC, Poppelaars F, Gaya da Costa M, et al. Distinct in vitro complement activation by various intravenous iron preparations. Am J Nephrol. 2017;45:49‐59.
    1. Rampton D, Folkersen J, Fishbane S, et al. Hypersensitivity reactions to intravenous iron: guidance for risk minimization and management. Haematologica. 2014;99:1671‐1676.
    1. Auerbach M, Ballard H, Glaspy J. Clinical update: intravenous iron for anaemia. Lancet. 2007;369:1502‐1504.
    1. Adkinson NF, Strauss WE, Bernard K, Kaper RF, Macdougall IC, Krop JS. Comparative safety of intravenous ferumoxytol versus ferric carboxymaltose for the treatment of iron deficiency anemia: rationale and study design of a randomized double‐blind study with a focus on acute hypersensitivity reactions. J Blood Med. 2017;8:155‐163.
    1. Adkinson NF, Strauss WE, Macdougall IC, et al. Comparative safety of intravenous ferumoxytol versus ferric carboxymaltose in iron deficiency anemia: a randomized trial. Am J Hematol. 2018;93:683‐690.
    1. Mordi IR, Tee A, Lang CC. Iron therapy in heart failure: ready for primetime. Card Fail Rev. 2018;4:28‐32.
    1. Jahn MR, Andreasen HB, Futterer S, et al. A comparative study of the physicochemical properties of iron isomaltoside 1000 (Monofer), a new intravenous iron preparation and its clinical implications. Eur J Pharm Biopharm. 2011;78:480‐491.
    1. Haase M, Bellomo R, Haase‐Fielitz A. Novel biomarkers, oxidative stress, and the role of labile iron toxicity in cardiopulmonary bypass‐associated acute kidney injury. J Am Coll Cardiol. 2010;55:2024‐2033.
    1. Panth N, Paudel KR, Parajuli K. Reactive oxygen species: a key hallmark of cardiovascular disease. Adv Med. 2016;2016:9152732.
    1. Hildebrandt PR, Bruun NE, Nielsen OW, et al. Effects of administration of iron isomaltoside 1000 in patients with chronic heart failure. A pilot study. Transfus Altern Transfus Med. 2010;11:131‐137.
    1. Wikstrom B, Bhandari S, Barany P, et al. Iron isomaltoside 1000: a new intravenous iron for treating iron deficiency in chronic kidney disease. J Nephrol. 2011;24:589‐596.
    1. Reinisch W, Altorjay I, Zsigmond F, et al. A 1‐year trial of repeated high‐dose intravenous iron isomaltoside 1000 to maintain stable hemoglobin levels in inflammatory bowel disease. Scand J Gastroenterol. 2015;50:1226‐1233.
    1. Reinisch W, Staun M, Tandon RK, et al. A randomized, open‐label, non‐inferiority study of intravenous iron isomaltoside 1000 (Monofer) compared with oral iron for treatment of anemia in IBD (PROCEED). Am J Gastroenterol. 2013;108:1877‐1888.
    1. Johansson PI, Rasmussen AS, Thomsen LL. Intravenous iron isomaltoside 1000 (Monofer) reduces postoperative anaemia in preoperatively non‐anaemic patients undergoing elective or subacute coronary artery bypass graft, valve replacement or a combination thereof: a randomized double‐blind placebo‐controlled clinical trial (the PROTECT trial). Vox Sang. 2015;109:257‐266.
    1. Bhandari S, Kalra PA, Kothari J, et al. A randomized, open‐label trial of iron isomaltoside 1000 (Monofer) compared with iron sucrose (Venofer) as maintenance therapy in haemodialysis patients. Nephrol Dial Transplant. 2015;30:1577‐1589.
    1. Kalra PA, Bhandari S, Saxena S, et al. A randomized trial of iron isomaltoside 1000 versus oral iron in non‐dialysis‐dependent chronic kidney disease patients with anaemia. Nephrol Dial Transplant. 2016;31:646‐655.
    1. Birgegard G, Henry D, Glaspy J, Chopra R, Thomsen LL, Auerbach M. A randomized, noninferiority trial of intravenous iron isomaltoside versus oral iron sulfate in patients with nonmyeloid malignancies and anemia receiving chemotherapy, the profound trial. Pharmacotherapy. 2016;36:402‐414.
    1. Dahlerup JF, Jacobsen BA, van der Woude J, Bark LA, Thomsen LL, Lindgren S. High‐dose fast infusion of parenteral iron isomaltoside is efficacious in inflammatory bowel disease patients with iron‐deficiency anaemia without profound changes in phosphate or fibroblast growth factor 23. Scand J Gastroenterol. 2016;51:1332‐1338.
    1. Holm C, Thomsen LL, Norgaard A, Langhoff‐Roos J. Single‐dose intravenous iron infusion or oral iron for treatment of fatigue after postpartum haemorrhage: a randomized controlled trial. Vox Sang. 2017;112:219‐228.
    1. Holm C, Thomsen LL, Norgaard A, Langhoff‐Roos J. Single‐dose intravenous iron infusion versus red blood cell transfusion for the treatment of severe postpartum anaemia: a randomized controlled pilot study. Vox Sang. 2017;112:122‐131.
    1. Gybel‐Brask M, Seeberg J, Thomsen LL, Johansson PI. Intravenous iron isomaltoside improves hemoglobin concentration and iron stores in female iron‐deficient blood donors: a randomized double‐blind placebo‐controlled clinical trial. Transfusion. 2018;58:974‐981.
    1. Derman R, Roman E, Modiano MR, Achebe MM, Thomsen LL, Auerbach M. A randomized trial of iron isomaltoside versus iron sucrose in patients with iron deficiency anemia. Am J Hematol. 2017;92:286‐291.
    1. Medical review of ferric carboxymaltose, application number 203565Orig1s000, Center for Drug Evaluation and Research, Food and Drug Administration, July 21, 2013. Food and Drug Administration. . Accessed May 22, 2019
    1. Wang C, Graham DJ, Kane RC, et al. Comparative risk of anaphylactic reactions associated with intravenous iron products. JAMA. 2015;314:2062‐2068.
    1. Kalra PA, Bhandari S. Safety of intravenous iron use in chronic kidney disease. Curr Opin Nephrol Hypertens. 2016;25:529‐535.
    1. Kalra PA, Bhandari S. Efficacy and safety of iron isomaltoside (Monofer) in the management of patients with iron deficiency anemia. Int J Nephrol Renovasc Dis. 2016;9:53‐64.
    1. Holm C, Thomsen LL, Langhoff‐Roos J. Intravenous iron isomaltoside treatment of women suffering from severe fatigue after postpartum hemorrhage. J Matern Fetal Neonatal Med. 2019;32:2797‐2804.

Source: PubMed

3
S'abonner