open access
Anemia in cancer patients: addressing a neglected issue - diagnostics and therapeutic algorithm


- Department of Oncology Diagnostics, Cardio-Oncology and Palliative Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Clinical Nutrition, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
open access
Abstract
Cancer-related anemia (CRA) continues to be a critical concern despite advancements in oncology treatments. The prevalence of anemia varies from 30% to 90%, impacting the quality of life and prognosis of cancer patients. While CRA is often attributed to antineoplastic therapies, it can also result from the disease itself. Inflammation and the iron regulatory hormone hepcidin play significant roles in CRA pathogenesis. Treatment-induced anemia caused by chemotherapy, tyrosine kinase inhibitors (TKIs) and immunotherapy, pose additional challenges. Intravenous (IV) iron has emerged as an effective treatment option for CRA, overcoming limitations associated with oral iron supplementation. Combining IV iron and ESAs enhances treatment outcomes. Future directions involve exploring ESA safety and their immunomodulatory effects. Transfusions provide quick relief but might impact prognosis and immune response. Other considerations include incorporating physical activity and exploring hepcidin-directed therapy. In conclusion, CRA management necessitates a multifaceted approach to address deficiencies, optimize therapies and improve patient outcomes.
Abstract
Cancer-related anemia (CRA) continues to be a critical concern despite advancements in oncology treatments. The prevalence of anemia varies from 30% to 90%, impacting the quality of life and prognosis of cancer patients. While CRA is often attributed to antineoplastic therapies, it can also result from the disease itself. Inflammation and the iron regulatory hormone hepcidin play significant roles in CRA pathogenesis. Treatment-induced anemia caused by chemotherapy, tyrosine kinase inhibitors (TKIs) and immunotherapy, pose additional challenges. Intravenous (IV) iron has emerged as an effective treatment option for CRA, overcoming limitations associated with oral iron supplementation. Combining IV iron and ESAs enhances treatment outcomes. Future directions involve exploring ESA safety and their immunomodulatory effects. Transfusions provide quick relief but might impact prognosis and immune response. Other considerations include incorporating physical activity and exploring hepcidin-directed therapy. In conclusion, CRA management necessitates a multifaceted approach to address deficiencies, optimize therapies and improve patient outcomes.
Keywords
anemia, cancer, hepcidin, erythropoiesis-stimulating agents, blood transfusions


Title
Anemia in cancer patients: addressing a neglected issue - diagnostics and therapeutic algorithm
Journal
Nowotwory. Journal of Oncology
Issue
Article type
Review paper
Published online
2023-09-12
Page views
21
Article views/downloads
13
DOI
Keywords
anemia
cancer
hepcidin
erythropoiesis-stimulating agents
blood transfusions
Authors
Konrad Tałasiewicz
Aleksandra Kapała


- Wojtukiewicz MZ, Sierko E, Rybaltowski M, et al. The Polish Cancer Anemia Survey (POLCAS): a retrospective multicenter study of 999 cases. Int J Hematol. 2009; 89(3): 276–284.
- Knight K, Wade S, Balducci L. Prevalence and outcomes of anemia in cancer: a systematic review of the literature. Am J Med. 2004; 116 Suppl 7A: 11S–26S.
- Harrison L, Shasha D, Shiaova L, et al. Prevalence of anemia in cancer patients undergoing radiation therapy. Semin Oncol. 2001; 28(2 Suppl 8): 54–59.
- Xu H, Xu L, Page JH, et al. Incidence of anemia in patients diagnosed with solid tumors receiving chemotherapy, 2010-2013. Clin Epidemiol. 2016; 8: 61–71.
- Cella D, Kallich J, McDermott A, et al. The longitudinal relationship of hemoglobin, fatigue and quality of life in anemic cancer patients: results from five randomized clinical trials. Ann Oncol. 2004; 15(6): 979–986.
- Barca-Hernando M, Muñoz-Martin AJ, Rios-Herranz E, et al. Case-Control Analysis of the Impact of Anemia on Quality of Life in Patients with Cancer: A Qca Study Analysis. Cancers (Basel). 2021; 13(11).
- Cappellini MD, Motta I. Anemia in Clinical Practice-Definition and Classification: Does Hemoglobin Change With Aging? Semin Hematol. 2015; 52(4): 261–269.
- National Cancer Institute, Cancer Therapy Evaluation Program Common Terminology Criteria for Adverse Events (CTCAE). https://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm#ctc_50 (24.07.2023).
- Gilreath JA, Rodgers GM. How I treat cancer-associated anemia. Blood. 2020; 136(7): 801–813.
- 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(7): 1886–1892.
- Park S, Jung CW, Kim K, et al. Iron deficient erythropoiesis might play key role in development of anemia in cancer patients. Oncotarget. 2015; 6(40): 42803–42812.
- Naoum FA. Iron deficiency in cancer patients. Rev Bras Hematol Hemoter. 2016; 38(4): 325–330.
- Gluszak C, de Vries-Brilland M, Seegers V, et al. Impact of Iron-Deficiency Management on Quality of Life in Patients with Cancer: A Prospective Cohort Study (CAMARA Study). Oncologist. 2022; 27(4): 328–333.
- Goodnough LT, Nemeth E, Ganz T. Detection, evaluation, and management of iron-restricted erythropoiesis. Blood. 2010; 116(23): 4754–4761.
- Gilreath JA, Stenehjem DD, Rodgers GM. Diagnosis and treatment of cancer-related anemia. Am J Hematol. 2014; 89(2): 203–212.
- Auerbach M, Ballard H, Trout JR, 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(7): 1301–1307.
- Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022; 12(1): 31–46.
- Macciò A, Madeddu C, Gramignano G, et al. The role of inflammation, iron, and nutritional status in cancer-related anemia: results of a large, prospective, observational study. Haematologica. 2015; 100(1): 124–132.
- Goodnough LT. Iron deficiency syndromes and iron-restricted erythropoiesis (CME). Transfusion. 2012; 52(7): 1584–1592.
- Ganz T. Anemia of Inflammation. N Engl J Med. 2019; 381(12): 1148–1157.
- Benjamin DJ, Xu A, Lythgoe MP, et al. Cancer Drug Approvals That Displaced Existing Standard-of-Care Therapies, 2016-2021. JAMA Netw Open. 2022; 5(3): e222265.
- Xu H, Xu L, Page JH, et al. Incidence of anemia in patients diagnosed with solid tumors receiving chemotherapy, 2010-2013. Clin Epidemiol. 2016; 8: 61–71.
- Barber NA, Afzal W, Akhtari M. Hematologic toxicities of small molecule tyrosine kinase inhibitors. Target Oncol. 2011; 6(4): 203–215.
- Kucharz J, Giza A, Dumnicka P, et al. Macrocytosis during sunitinib treatment predicts progression-free survival in patients with metastatic renal cell carcinoma. Med Oncol. 2016; 33(10): 109.
- Schallier D, Trullemans F, Fontaine C, et al. Tyrosine kinase inhibitor-induced macrocytosis. Anticancer Res. 2009; 29(12): 5225–5228.
- Kloth JSL, Hamberg P, Mendelaar PAJ, et al. Macrocytosis as a potential parameter associated with survival after tyrosine kinase inhibitor treatment. Eur J Cancer. 2016; 56: 101–106.
- Dores GM, Nayernama A, Cheng C, et al. Hemolytic anemia following alectinib reported to the U.S. Food and Drug Administration Adverse Event Reporting System. Am J Hematol. 2022; 97(4): E129–E132.
- Kroll MH, Rojas-Hernandez C, Yee C. Hematologic complications of immune checkpoint inhibitors. Blood. 2022; 139(25): 3594–3604.
- Guo Q, Zhao JN, Liu T, et al. Immune checkpoint inhibitor-induced aplastic anaemia: Case series and large-scale pharmacovigilance analysis. Front Pharmacol. 2023; 14: 1057134.
- Rund D. Intravenous iron: do we adequately understand the short- and long-term risks in clinical practice? Br J Haematol. 2021; 193(3): 466–480.
- Moretti D, Goede JS, Zeder C, et al. Oral iron supplements increase hepcidin and decrease iron absorption from daily or twice-daily doses in iron-depleted young women. Blood. 2015; 126(17): 1981–1989.
- Stoffel NU, Cercamondi CI, Brittenham G, et al. Iron absorption from oral iron supplements given on consecutive versus alternate days and as single morning doses versus twice-daily split dosing in iron-depleted women: two open-label, randomised controlled trials. Lancet Haematol. 2017; 4(11): e524–e533.
- Aapro M, Beguin Y, Bokemeyer C, et al. ESMO Guidelines Committee. Management of anaemia and iron deficiency in patients with cancer: ESMO Clinical Practice Guidelines. Ann Oncol. 2018; 29(Suppl 4): iv96–iv9iv110.
- Radziwon P, Krzakowski M, Kalinka-Warzocha E, et al. Anemia in cancer patients — Expert Group recommendations. Oncol Clin Pract. 2017; 13: 202–210.
- Makharadze T, Boccia R, Krupa A, et al. Efficacy and safety of ferric carboxymaltose infusion in reducing anemia in patients receiving chemotherapy for nonmyeloid malignancies: A randomized, placebo-controlled study (IRON-CLAD). Am J Hematol. 2021; 96(12): 1639–1646.
- Jang JHo, Kim Y, Park S, et al. Efficacy of intravenous iron treatment for chemotherapy-induced anemia: A prospective Phase II pilot clinical trial in South Korea. PLoS Med. 2020; 17(6): e1003091.
- Luporsi E, Mahi L, Morre C, et al. Evaluation of cost savings with ferric carboxymaltose in anemia treatment through its impact on erythropoiesis-stimulating agents and blood transfusion: French healthcare payer perspective. J Med Econ. 2012; 15(2): 225–232.
- Gluszak C, de Vries-Brilland M, Seegers V, et al. Impact of Iron-Deficiency Management on Quality of Life in Patients with Cancer: A Prospective Cohort Study (CAMARA Study). Oncologist. 2022; 27(4): 328–333.
- GOETSCH A, MOORE C, MINNICH V. OBSERVATIONS ON THE EFFECT OF MASSIVE DOSES OF IRON GIVEN INTRAVENOUSLY TO PATIENTS WITH HYPOCHROMIC ANEMIA. Blood. 1946; 1(2): 129–142.
- 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(15): 2293–2306.
- Wysowski DK, Swartz L, Borders-Hemphill BV, et al. Use of parenteral iron products and serious anaphylactic-type reactions. Am J Hematol. 2010; 85(9): 650–654.
- Avni T, Bieber A, Grossman A, et al. The safety of intravenous iron preparations: systematic review and meta-analysis. Mayo Clin Proc. 2015; 90(1): 12–23.
- European Medicines Agency. New recommendations to manage risk of allergic reactions with intravenous iron-containing medicines 2013. http://www.ema.europa.eu/docs/en_GB/document_library/Press_release/2013/06/WC500144874.pdf (27.07.2023).
- Rampton D, Folkersen J, Fishbane S, et al. Hypersensitivity reactions to intravenous iron: guidance for risk minimization and management. Haematologica. 2014; 99(11): 1671–1676.
- Lim W, Afif W, Knowles S, et al. Canadian expert consensus: management of hypersensitivity reactions to intravenous iron in adults. Vox Sang. 2019; 114(4): 363–373.
- Litton E, Xiao J, Ho KM. Safety and efficacy of intravenous iron therapy in reducing requirement for allogeneic blood transfusion: systematic review and meta-analysis of randomised clinical trials. BMJ. 2013; 347: f4822.
- Shah A, Fisher SA, Wong H, et al. Safety and efficacy of iron therapy on reducing red blood cell transfusion requirements and treating anaemia in critically ill adults: a systematic review with meta-analysis and trial sequential analysis. J Crit Care. 2019; 49: 162–171.
- Lin FK, Suggs S, Lin CH, et al. Cloning and expression of the human erythropoietin gene. Proc Natl Acad Sci U S A. 1985; 82(22): 7580–7584.
- Schoen MW, Hoque S, Witherspoon BJ, et al. End of an era for erythropoiesis-stimulating agents in oncology. Int J Cancer. 2020; 146(10): 2829–2835.
- Bennett CL, Silver SM, Djulbegovic B, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA. 2008; 299(8): 914–924.
- Gergal Gopalkrishna Rao SR, Bugazia S, Dhandapani TP, et al. Efficacy and Cardiovascular Adverse Effects of Erythropoiesis Stimulating Agents in the Treatment of Cancer-Related Anemia: A Systematic Review of Randomized Controlled Trials. Cureus. 2021; 13(9): e17835.
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Hematopoietic Growth Factors Version 2.2023 — March 6, 2023. © National Comprehensive CancerNetwork, Inc. 2023. All rights reserved. Accessed 27 July 2023. To view the most recent and complete version of the guideline, go online to NCCN.org.
- Khorana AA, Kuderer NM, Culakova E, et al. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood. 2008; 111(10): 4902–4907.
- Brar SK, Perveen S, Chaudhry MR, et al. Erythropoietin-Induced Hypertension: A Review of Pathogenesis, Treatment, and Role of Blood Viscosity. Cureus. 2021; 13(1): e12804.
- Kumar SM, Zhang G, Bastian BC, et al. Erythropoietin and erythropoietin receptor expression in human cancer. Cancer Res. 2001; 61(9): 3561–3565.
- 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(9): 1027–1032.
- 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(25): 5960–5972.
- Aapro M, Moebus V, Nitz U, et al. Safety and efficacy outcomes with erythropoiesis-stimulating agents in patients with breast cancer: a meta-analysis. Ann Oncol. 2015; 26(4): 688–695.
- Vansteenkiste J, Glaspy J, Henry D, et al. Benefits and risks of using erythropoiesis-stimulating agents (ESAs) in lung cancer patients: study-level and patient-level meta-analyses. Lung Cancer. 2012; 76(3): 478–485.
- Glaspy J, Crawford J, Vansteenkiste J, et al. Erythropoiesis-stimulating agents in oncology: a study-level meta-analysis of survival and other safety outcomes. Br J Cancer. 2010; 102(2): 301–315.
- Ludwig H, Crawford J, Osterborg A, et al. Pooled analysis of individual patient-level data from all randomized, double-blind, placebo-controlled trials of darbepoetin alfa in the treatment of patients with chemotherapy-induced anemia. J Clin Oncol. 2009; 27(17): 2838–2847.
- Gascón P, Nagarkar R, Šmakal M, et al. A Randomized, Double-Blind, Placebo-Controlled, Phase III Noninferiority Study of the Long-Term Safety and Efficacy of Darbepoetin Alfa for Chemotherapy-Induced Anemia in Patients With Advanced NSCLC. J Thorac Oncol. 2020; 15(2): 190–202.
- Bastit L, Vandebroek An, Altintas S, et al. Randomized, multicenter, controlled trial comparing the efficacy and safety of darbepoetin alpha administered every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. J Clin Oncol. 2008; 26(10): 1611–1618.
- Silva I, Alípio C, Pinto R, et al. Potential anti-inflammatory effect of erythropoietin in non-clinical studies in vivo: A systematic review. Biomed Pharmacother. 2021; 139: 111558.
- 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(8): 2197–2206.
- Schrijvers D. Management of anemia in cancer patients: transfusions. Oncologist. 2011; 16 Suppl 3: 12–18.
- Chau JKM, Harris JR, Seikaly HR. Transfusion as a predictor of recurrence and survival in head and neck cancer surgery patients. J Otolaryngol Head Neck Surg. 2010; 39(5): 516–522.
- Anic K, Schmidt MW, Schmidt M, et al. Impact of perioperative red blood cell transfusion, anemia of cancer and global health status on the prognosis of elderly patients with endometrial and ovarian cancer. Front Oncol. 2022; 12: 967421.
- Al-Refaie WB, Parsons HM, Markin A, et al. Blood transfusion and cancer surgery outcomes: a continued reason for concern. Surgery. 2012; 152(3): 344–354.
- Busch OR, Hop WC, Hoynck van Papendrecht MA, et al. Blood transfusions and prognosis in colorectal cancer. N Engl J Med. 1993; 328(19): 1372–1376.
- Squires MH, Kooby DA, Poultsides GA, et al. Effect of Perioperative Transfusion on Recurrence and Survival after Gastric Cancer Resection: A 7-Institution Analysis of 765 Patients from the US Gastric Cancer Collaborative. J Am Coll Surg. 2015; 221(3): 767–777.
- Kooby DA, Stockman J, Ben-Porat L, et al. Influence of transfusions on perioperative and long-term outcome in patients following hepatic resection for colorectal metastases. Ann Surg. 2003; 237(6): 860–9; discussion 869.
- Khorana AA, Francis CW, Blumberg N, et al. Blood transfusions, thrombosis, and mortality in hospitalized patients with cancer. Arch Intern Med. 2008; 168(21): 2377–2381.
- Xenos ES, Vargas HD, Davenport DL. Association of blood transfusion and venous thromboembolism after colorectal cancer resection. Thromb Res. 2012; 129(5): 568–572.
- Vamvakas EC, Blajchman MA. Deleterious clinical effects of transfusion-associated immunomodulation: fact or fiction? Blood. 2001; 97(5): 1180–1195.
- Cata JP, Wang H, Gottumukkala V, et al. Inflammatory response, immunosuppression, and cancer recurrence after perioperative blood transfusions. Br J Anaesth. 2013; 110(5): 690–701.
- Mispelbaum R, Hattenhauer ST, Brossart P, et al. Red blood cell transfusions impact response rates to immunotherapy in patients with solid malignant tumors. Front Immunol. 2022; 13: 976011.
- Carson JL, Stanworth SJ, Dennis JA, et al. Transfusion thresholds for guiding red blood cell transfusion. Cochrane Database Syst Rev. 2021; 12(12): CD002042.
- Watkins T, Surowiecka MK, McCullough J. Transfusion indications for patients with cancer. Cancer Control. 2015; 22(1): 38–46.
- Scott JM, Weir DG. Drug-induced megaloblastic change. Clin Haematol. 1980; 9(3): 587–606.
- Danaei G, Vander Hoorn S, Lopez AD, et al. Comparative Risk Assessment collaborating group (Cancers). Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet. 2005; 366(9499): 1784–1793.
- Dong B, Qi Y, Lin Lu, et al. Which Exercise Approaches Work for Relieving Cancer-Related Fatigue? A Network Meta-analysis. J Orthop Sports Phys Ther. 2023; 0(6): 1–10.
- Brown JC, Ma C, Shi Q, et al. Association between physical activity and the time course of cancer recurrence in stage III colon cancer. Br J Sports Med. 2023; 57(15): 965–971.
- Rørth M, Madsen KR, Burmølle SH, et al. Effects of Darbepoetin Alfa with exercise in cancer patients undergoing chemotherapy: an explorative study. Scand J Med Sci Sports. 2011; 21(3): 369–377.
- Drouin JS, Young TJ, Beeler J, et al. Random control clinical trial on the effects of aerobic exercise training on erythrocyte levels during radiation treatment for breast cancer. Cancer. 2006; 107(10): 2490–2495.
- Mohamady HM, Elsisi HF, Aneis YM. Impact of moderate intensity aerobic exercise on chemotherapy-induced anemia in elderly women with breast cancer: A randomized controlled clinical trial. J Adv Res. 2017; 8(1): 7–12.
- Ashem HN, Hamada HA, Abbas RL. Effect of aerobic exercise on immunoglobulins and anemia after chemotherapy in breast cancer patients. J Bodyw Mov Ther. 2020; 24(3): 137–140.
- Vadhan-Raj S, Abonour R, Goldman JW, et al. A first-in-human phase 1 study of a hepcidin monoclonal antibody, LY2787106, in cancer-associated anemia. J Hematol Oncol. 2017; 10(1): 73.
- Jeng SS, Chen YH. Association of Zinc with Anemia. Nutrients. 2022; 14(22).
- Greffeuille V, Fortin S, Gibson R, et al. Associations between Zinc and Hemoglobin Concentrations in Preschool Children and Women of Reproductive Age: An Analysis of Representative Survey Data from the Biomarkers Reflecting Inflammation and Nutritional Determinants of Anemia (BRINDA) Project. J Nutr. 2021; 151(5): 1277–1285.