Treatment of anemia in cancer patients undergoing chemotherapy with intravenous ferric carboxymaltose without erythropoiesis-stimulating agents

Hikmat Abdel-Razeq, Salwa S Saadeh, Razan Malhis, Sameer Yasser, Hazem Abdulelah, Rana Eljaber, Amer Kleib, Rouba Ismael, Hikmat Abdel-Razeq, Salwa S Saadeh, Razan Malhis, Sameer Yasser, Hazem Abdulelah, Rana Eljaber, Amer Kleib, Rouba Ismael

Abstract

Background: Anemia is commonly encountered in cancer patients receiving active chemotherapy. Due to adverse events and presumed negative effects on disease-progression and survival, erythropoiesis-stimulating agents are not frequently used. In this study, we assess the efficacy and safety of intravenous ferric carboxymaltose (FCM) to treat cancer-induced anemia (CIA).

Patients and methods: We recruited adult cancer patients on active chemotherapy with a hemoglobin (Hb) level ⩽11.0 g/dL. Based on serum ferritin (sFr) and transferrin saturation (TSAT), patients were divided into 3 groups: group I (absolute iron deficiency, n = 26) with sFr < 30 ng/mL and TSAT < 20%; group II (functional iron deficiency, n = 24) with sFr 30-800 ng/mL and TSAT < 20%; and patients with TSAT ⩾ 20% were placed in group III as "others" (n = 34). All patients were treated with intravenous FCM. Serum hepcidin and C-reactive protein were used as biomarkers to predict response.

Results: A total of 84 patients with a median age (SD) of 53.8 (10.6) were recruited. Baseline median Hb level was 10.2 (range: 8.3-11.0) gm/dL. At week 12, there was a significant increment in Hb level for patients in groups I and II (median increment: 2.35 and 1.5 gm/dL, respectively), with limited response observed in group III, and most of the increment noted as early as week 3 (⩾1.0 g/dL). Responders tended to have lower levels of hepcidin. No clinically significant adverse events were reported; however, asymptomatic hypophosphatemia was observed in 39 (46.4%) patients.

Conclusions: Intravenous FCM is a safe and effective treatment option for the management of a subgroup of patients with CIA.The study was registered at ClinicalTrials.gov [Identifier: NCT04246021].

Keywords: anemia; cancer; ferric carboxymaltose; hepcidin.

Conflict of interest statement

Conflict of interest statement: The authors declare that there is no conflict of interest.

© The Author(s), 2020.

Figures

Figure 1.
Figure 1.
Hemoglobin changes following intravenous iron infusion.

References

    1. Ludwig H, Van Belle S, Barrett-Lee P, et al. The European cancer anaemia survey (ECAS): a large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur J Cancer 2004; 40: 2293–2306.
    1. Ludwig H, Müldür E, Endler G, et al. Prevalence of iron deficiency across different tumors and its association with poor performance status, disease status and anemia. Ann Oncol 2013; 24: 1886–1892.
    1. Grotto H. Anaemia of cancer: an overview of mechanisms involved in its pathogenesis. Med Oncol 2007; 25: 12–21.
    1. Abdel-Razeq HN. Cancer-related anemia. Saudi Med J 2004; 25: 15–20.
    1. Crawford J, Cella D, Cleeland C, et al. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002; 95: 888–895.
    1. Thomas G. The effect of hemoglobin level on radiotherapy outcomes: the Canadian experience. Semin Oncol 2001; 28: 60–65.
    1. Connor J, O’Shea A, McCool K, et al. Peri-operative allogeneic blood transfusion is associated with poor overall survival in advanced epithelial ovarian cancer; potential impact of patient blood management on cancer outcomes. Gynecol Oncol 2018; 151: 294–298.
    1. Gordon D, Nichols G, Ben-Jacob A, et al. Treating anemia of cancer with every-4-week darbepoetin alfa: final efficacy and safety results from a phase II, randomized, double-blind, placebo-controlled study. Oncologist 2008; 13: 715–724.
    1. Ferrario E, Ferrari L, Bidoli P, et al. Treatment of cancer-related anemia with epoetin alfa: a review. Cancer Treat Rev 2004; 30: 563–575.
    1. Littlewood T, Bajetta E, Nortier J, et al. Effects of epoetin alfa on hematologic parameters and quality of life in cancer patients receiving nonplatinum chemotherapy: results of a randomized, double-blind, placebo-controlled trial. J Clin Oncol 2001; 19: 2865–2874.
    1. Gabrilove J, Cleeland C, Livingston R, et al. Clinical evaluation of once-weekly dosing of epoetin alfa in chemotherapy patients: improvements in hemoglobin and quality of life are similar to three-times-weekly dosing. J Clin Oncol 2001; 19: 2875–2882.
    1. Aapro M, Österborg A, Gascón P, et al. Prevalence and management of cancer-related anaemia, iron deficiency and the specific role of i.v. iron. Ann Oncol 2012; 23: 1954–1962.
    1. Auerbach M, Ballard H, Trout J, et al. Intravenous iron optimizes the response to recombinant human erythropoietin in cancer patients with chemotherapy-related anemia: a multicenter, open-label, randomized trial. J Clin Oncol 2004; 22: 1301–1307.
    1. Auerbach M, Silberstein P, Webb R, et al. Darbepoetin alfa 300 or 500 μg once every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. Am J Hematol 2010; 85: 655–663.
    1. Henry D, Dahl N, Auerbach M, et al. Intravenous ferric gluconate significantly improves response to epoetin alfa versus oral iron or no iron in anemic patients with cancer receiving chemotherapy. Oncologist 2007; 12: 231–242.
    1. Bastit L, Vandebroek A, Altintas S, et al. Randomized, multicenter, controlled trial comparing the efficacy and safety of darbepoetin alfa administered every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. J Clin Oncol 2008; 26: 1611–1618.
    1. Henke M, Laszig R, Rübe C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. Lancet 2003; 362: 1255–1260.
    1. Leyland-Jones B, Semiglazov V, Pawlicki M, et al. Maintaining normal hemoglobin levels with epoetin alfa in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. J Clin Oncol 2005; 23: 5960–5972.
    1. Wright JR, Ung YC, Julian JA, et al. Randomized, double-blind, placebo-controlled trial of erythropoietin in non–small-cell lung cancer with disease-related anemia. J Clin Oncol 2007; 25: 1027–1032.
    1. Bennett CL. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA 2008; 299: 914.
    1. Khorana AA, Francis CW, Blumberg N, et al. Blood transfusions, thrombosis, and mortality in hospitalized patients with cancer. Arch Intern Med 2008; 168: 2377.
    1. Rizzo JD, Brouwers M, Hurley P, et al. American society of clinical oncology/American society of hematology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. J Oncol Pract 2010; 6: 317–320.
    1. Abdel-Razeq H, Hijjawi S, Abdulelah H, et al. The impact of recently published negative erythropoiesis-stimulating agent studies on the clinical management of cancer-related anemia at a single center. Hematol Oncol Stem Cell Res 2010; 3: 78–83.
    1. Ludwig H, Aapro M, Bokemeyer C, et al. A European patient record study on diagnosis and treatment of chemotherapy-induced anaemia. Support Care Cancer 2014; 22: 2197–2206.
    1. Onken JE, Bregman DB, Harrington RA, et al. A multicenter, randomized, active-controlled study to investigate the efficacy and safety of intravenous ferric carboxymaltose in patients with iron deficiency anemia. Transfusion. Epub ahead of print 17 June 2013. DOI: 10.1111/trf.12289.
    1. Qunibi WY, Martinez C, Smith M, et al. A randomized controlled trial comparing intravenous ferric carboxymaltose with oral iron for treatment of iron deficiency anaemia of non-dialysis-dependent chronic kidney disease patients. Nephrol Dial Transplant 2010; 26: 1599–1607.
    1. Thomas C, Thomas L. Biochemical markers and hematologic indices in the diagnosis of functional iron deficiency. Clin Chem 2002; 48: 1066–1076.
    1. Sheetz M, Barrington P, Callies S, et al. Targeting the hepcidin-ferroportin pathway in anaemia of chronic kidney disease. Br J Clin Pharmacol 2019; 85: 935–948.
    1. Stoffel NU, Lazrak M, Bellitir S, et al. The opposing effects of acute inflammation and iron deficiency anemia on serum hepcidin and iron absorption in young women. Haematologica 2019; 104: 1143–1149.
    1. Arezes J, Nemeth E. Hepcidin and iron disorders: new biology and clinical approaches. Int J Lab Hematol 2015; 37: 92–98.
    1. Ruchala P, Nemeth E. The pathophysiology and pharmacology of hepcidin. Trends Pharmacol Sci 2014; 35: 155–161.
    1. Camaschella C. Iron and hepcidin: a story of recycling and balance. Hematology 2013; 2013: 1–8.
    1. Sangkhae V, Nemeth E. Regulation of the iron homeostatic hormone hepcidin. Adv Nutr 2017; 8: 126–136.
    1. Abdel-Razeq H, Abbasi S, Saadi I, et al. Intravenous iron monotherapy for the treatment of non-iron-deficiency anemia in cancer patients undergoing chemotherapy: a pilot study. Drug Des Devel Ther 2013; 7: 939–944.
    1. Wyck DBV, Mangione A, Morrison J, et al. Large-dose intravenous ferric carboxymaltose injection for iron deficiency anemia in heavy uterine bleeding: a randomized, controlled trial. Transfusion 2009; 49: 2719–2728.
    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. Bager P, Hvas CL, Dahlerup JF. Drug-specific hypophosphatemia and hypersensitivity reactions following different intravenous iron infusions. Br J Clin Pharmacol 2017; 83: 1118–1125.

Source: PubMed

3
Předplatit