665
Views
8
CrossRef citations to date
0
Altmetric
Review

Experimental Drugs for Chemotherapy- and Cancer-Related Anemia

, ORCID Icon, , &
Pages 593-611 | Published online: 24 Jun 2021

References

  • McLean E, Cogswell M, Egli I, Wojdyla D, de Benoist B. Worldwide prevalence of anaemia, WHO vitamin and mineral nutrition information system, 1993–2005. Public Health Nutr. 2009;12(4):444–454. doi:10.1017/S1368980008002401
  • Madeddu C, Gramignano G, Astara G, et al. Pathogenesis and treatment options of cancer related anemia: perspective for a targeted mechanism-based approach. Front Physiol. 2018;9:1294. doi:10.3389/fphys.2018.01294
  • Birgegård G, Aapro MS, Bokemeyer C, et al. Cancer-related anemia: pathogenesis, prevalence and treatment. Oncology. 2005;68(Suppl 1):3–11. doi:10.1159/000083128
  • Schwartz RN. Anemia in patients with cancer: incidence, causes, impact, management, and use of treatment guidelines and protocols. Am J Health Syst Pharm. 2007;64(3Suppl 2):S5–S13;quiz S28–S30. doi:10.2146/ajhp060601
  • Wu Y, Aravind S, Ranganathan G, Martin A, Nalysnyk L. Anemia and thrombocytopenia in patients undergoing chemotherapy for solid tumors: a descriptive study of a large outpatient oncology practice database, 2000–2007. Clin Ther. 2009;31(Pt 2):2416–2432. doi:10.1016/j.clinthera.2009.11.020
  • Knight K, Wade S, Balducci L. Prevalence and outcomes of anemia in cancer: a systematic review of the literature. Am J Med. 2004;116(Suppl7A):11S–26S. doi:10.1016/j.amjmed.2003.12.008
  • Melo-Alvim C, Miguel-Semedo P, Paiva RS, et al. Pretreatment hemoglobin level as a prognostic factor in patients with locally advanced head and neck squamous cell carcinoma. Rep Pract Oncol Radiother. 2020;25(5):768–774. doi:10.1016/j.rpor.2020.07.002
  • An MS, Yoo JH, Kim KH, et al. T4 stage and preoperative anemia as prognostic factors for the patients with colon cancer treated with adjuvant FOLFOX chemotherapy. World J Surg Oncol. 2015;13:64. doi:10.1186/s12957-015-0488-7
  • Shin NR, Lee YY, Kim SH, et al. Prognostic value of pretreatment hemoglobin level in patients with early cervical cancer. Obstet Gynecol Sci. 2014;57(1):28–36. doi:10.5468/ogs.2014.57.1.28
  • Zhang Y, Chen Y, Chen D, et al. Impact of preoperative anemia on relapse and survival in breast cancer patients. BMC Cancer. 2014;14:844. doi:10.1186/1471-2407-14-844
  • Obermair A, Petru E, Windbichler G, et al. Significance of pretreatment serum hemoglobin and survival in epithelial ovarian cancer. Oncol Rep. 2000;7:639–644. doi:10.3892/or.7.3.639
  • Lee WR, Berkey B, Marcial V, et al. Anemia is associated with decreased survival and increased locoregional failure in patients with locally advanced head and neck carcinoma: a secondary analysis of RTOG 85-27. Int J Radiat Oncol Biol Phys. 1998;42(5):1069–1075. doi:10.1016/S0360-3016(98)00348-4
  • Grant DG, Hussain A, Hurman D. Pre-treatment anaemia alters outcome in early squamous cell carcinoma of the larynx treated by radical radiotherapy. J Laryngol Otol. 1999;113(9):829–833. doi:10.1017/S0022215100145323
  • Prosnitz RG, Yao B, Farrell CL, Clough R, Brizel DM. Pretreatment anemia is correlated with the reduced effectiveness of radiation and concurrent chemotherapy in advanced head and neck cancer. Int J Radiat Oncol Biol Phys. 2005;61(4):1087–1095. doi:10.1016/j.ijrobp.2004.07.710
  • Fuso L, Mazzola S, Marocco F, et al. Pretreatment serum hemoglobin level as a predictive factor of response to neoadjuvant chemotherapy in patients with locally advanced squamous cervical carcinoma: a preliminary report. Gynecol Oncol. 2005;99(3 Suppl 1):S187–S191. doi:10.1016/j.ygyno.2005.07.079
  • Zhu W, Xu B. Association of pretreatment anemia with pathological response and survival of breast cancer patients treated with neoadjuvant chemotherapy: a population-based study. PLoS One. 2015;10(8):e0136268. doi:10.1371/journal.pone.0136268
  • Harris AL. Hypoxia-a key regulatory factor in tumour growth. Nat Rev Cancer. 2002;2(1):38–47. doi:10.1038/nrc704
  • Holzner B, Kemmler G, Greil R, et al. The impact of hemoglobin levels on fatigue and quality of life in cancer patients. Ann Oncol. 2002;13(6):965–973. doi:10.1093/annonc/mdf122
  • Ludwig H, Müldür E, Endler G, Hübl W. 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. doi:10.1093/annonc/mdt118
  • Owusu C, Cohen HJ, Feng T, et al.; Cancer and Aging Research Group (CARG). Anemia and functional disability in older adults with cancer. J Natl Compr Canc Netw. 2015;13(10):1233–1239. doi:10.6004/jnccn.2015.0152
  • Spivak JL. Cancer-related anemia: its causes and characteristics. Semin Oncol. 1994;21(2 Suppl 3):3–8.
  • Adamson JW. The anemia of inflammation/malignancy: mechanisms and management. Hematol Am Soc Hematol Educ Program. 2008;2008(1):159–165. doi:10.1182/asheducation-2008.1.159
  • Spivak JL. The anaemia of cancer: death by a thousand cuts. Nat Rev Cancer. 2005;5(7):543–555. doi:10.1038/nrc1648
  • Miller CB, Jones RJ, Piantadosi S, Abeloff MD, Spivak JL. Decreased erythropoietin response in patients with the anemia of cancer. N Engl J Med. 1990;322(24):1689–1692. doi:10.1056/NEJM199006143222401
  • Aapro M, Österborg A, Gascón P, Ludwig H, Beguin Y. Prevalence and management of cancer-related anaemia, iron deficiency and the specific role of i.v. iron. Ann Oncol. 2012;23(8):1954–1962. doi:10.1093/annonc/mds112
  • National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Hematopoietic Growth Factors. Version 2; March 23, 2021. Available from: https://www.nccn.org/professionals/physician_gls/pdf/growthfactors.pdf. Accessed April 13, 2021.
  • Spivak JL. Iron and the anemia of chronic disease. Oncology (Huntingt). 2002;16(9 Suppl 10):25–33.
  • Faquin WC, Schneider TJ, Goldberg MA. Effect of inflammatory cytokines on hypoxia-induced erythropoietin production. Blood. 1992;79(8):1987–1994. doi:10.1182/blood.V79.8.1987.1987
  • Jewell UR, Kvietikova I, Scheid A, Bauer C, Wenger RH, Gassmann M. Induction of HIF-1 alpha in response to hypoxia is instantaneous. FASEB J. 2001;15(7):1312–1314. doi:10.1096/fj.00-0732fje
  • Lang F, Abed M, Lang E, Föller M. Oxidative stress and suicidal erythrocyte death. Antioxid Redox Signal. 2014;21(1):138–153. doi:10.1089/ars.2013.5747
  • 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. doi:10.3324/haematol.2014.112813
  • Andrews NC. Anemia of inflammation: the cytokine-hepcidin link. J Clin Invest. 2004;113(9):1251–1253. doi:10.1172/JCI21441
  • Ganz T. Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation. Blood. 2003;102(3):783–788. doi:10.1182/blood-2003-03-0672
  • Macciò A, Madeddu C, Gramignano G, et al. A randomized phase III clinical trial of a combined treatment for cachexia in patients with gynecological cancers: evaluating the impact on metabolic and inflammatory profiles and quality of life. Gynecol Oncol. 2012;124(3):417–425. doi:10.1016/j.ygyno.2011.12.435
  • U.S. Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE). Version 5.0; November 27, 2017. Available from: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_5x7.pdf. Accessed February 02, 2021.
  • Dalton JD, Bailey NP, Barrett-Lee PJ, O’Brien MER. Multicenter UK audit of anemia in patients receiving cytotoxic chemotherapy [abstract]. Proc ASCO. 1998;17:418a.
  • Cheng K, Zhao F, Gao F, et al. Factors potentially associated with chemotherapy-induced anemia in patients with solid cancers. Asian Pac J Cancer Prev. 2012;13(10):5057–5061. doi:10.7314/APJCP.2012.13.10.5057
  • Groopman JE, Itri LM. Chemotherapy-induced anemia in adults: incidence and treatment. J Natl Cancer Inst. 1999;91(19):1616–1634. doi:10.1093/jnci/91.19.1616
  • Perazella MA, Moeckel GW. Nephrotoxicity from chemotherapeutic agents: clinical manifestations, pathobiology, and prevention/therapy. Semin Nephrol. 2010;30(6):570–581. doi:10.1016/j.semnephrol.2010.09.005
  • Neumcke I, Schneider B, Fandrey J, Pagel H. Effects of pro- and antioxidative compounds on renal production of erythropoietin. Endocrinology. 1999;140(2):641–645. doi:10.1210/endo.140.2.6529
  • Macciò A, Madeddu C. Cisplatin: an old drug with a newfound efficacy - from mechanisms of action to cytotoxicity. Expert Opin Pharmacother. 2013;14(13):1839–1857. doi:10.1517/14656566.2013.813934
  • Barni S, Cabiddu M, Guarneri P, Lonati V, Petrelli F. The risk for anemia with targeted therapies for solid tumors. Oncologist. 2012;17(5):715–724. doi:10.1634/theoncologist.2012-0024
  • Ruiz-Schutz VC, Gomes LM, Mariano RC, et al. Risk of fatigue and anemia in patients with advanced cancer treated with olaparib: a meta-analysis of randomized controlled trials. Crit Rev Oncol Hematol. 2019;141:163–173. doi:10.1016/j.critrevonc.2019.06.012
  • Moore KN, Mirza MR, Matulonis UA. The poly (ADP ribose) polymerase inhibitor niraparib: management of toxicities. Gynecol Oncol. 2018;149(1):214–220. doi:10.1016/j.ygyno.2018.01.011
  • Wahlberg E, Karlberg T, Kouznetsova E, et al. Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitors. Nat Biotechnol. 2012;30(3):283–288. doi:10.1038/nbt.2121
  • Farrés J, Martín-Caballero J, Martínez C, et al. Parp-2 is required to maintain hematopoiesis following sublethal γ-irradiation in mice. Blood. 2013;122(1):44–54. doi:10.1182/blood-2012-12-472845
  • Farrés J, Llacuna L, Martin-Caballero J, et al. PARP-2 sustains erythropoiesis in mice by limiting replicative stress in erythroid progenitors. Cell Death Differ. 2015;22(7):1144–1157. doi:10.1038/cdd.2014.202
  • Pelham C, Jimenez T, Rodova M, Rudolph A, Chipps E, Islam MR. Regulation of HFE expression by poly(ADP-ribose) polymerase-1 (PARP1) through an inverted repeat DNA sequence in the distal promoter. Biochim Biophys Acta. 2013;1829(12):1257–1265. doi:10.1016/j.bbagrm.2013.10.002
  • Gao F, Cheng K, Zhao F, et al. Prevalence and characteristics of anemia in patients with solid cancers at diagnosis in southwest China. Asian Pac J Cancer Prev. 2011;12(11):2825–2828.
  • Ganz T. Anemia of inflammation. N Engl J Med. 2019;381(12):1148–1157. doi:10.1056/NEJMra1804281
  • Macciò A, Madeddu C, Massa D, et al. Hemoglobin levels correlate with interleukin-6 levels in patients with advanced untreated epithelial ovarian cancer: role of inflammation in cancer-related anemia. Blood. 2005;106(1):362–367. doi:10.1182/blood-2005-01-0160
  • Gilreath JA, Rodgers GM. How I treat cancer-associated anemia. Blood. 2020;136(7):801–813. doi:10.1182/blood.2019004017
  • Proctor MJ, Morrison DS, Talwar D, et al. An inflammation-based prognostic score (mGPS) predicts cancer survival independent of tumour site: a Glasgow Inflammation Outcome Study. Br J Cancer. 2011;104(4):726–734. doi:10.1038/sj.bjc.6606087
  • Crawford J, Cella D, Cleeland CS, 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(4):888–895. doi:10.1002/cncr.10763
  • Davies KJ, Maguire JJ, Brooks GA, Dallman PR, Packer L. Muscle mitochondrial bioenergetics, oxygen supply, and work capacity during dietary iron deficiency and repletion. Am J Physiol. 1982;242(6):E418–E427. doi:10.1152/ajpendo.1982.242.6.E418
  • Poulos TL. Heme enzyme structure and function. Chem Rev. 2014;114(7):3919–3962.
  • Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med. 2005;352(10):1011–1023. doi:10.1056/NEJMra041809
  • Carson JL, Grossman BJ, Kleinman S, et al. Red blood cell transfusion: a clinical practice guideline from the AABB. Ann Intern Med. 2012;157(1):49–58. doi: 10.7326/0003-4819-157-1-201206190-00429
  • Mercadante S, Ferrara P, Villari P, David F, Giarratano A, Riina S. Effects of red blood transfusion on anemia-related symptoms in patients with cancer. J Palliat Med. 2009;12(1):60–63. doi:10.1089/jpm.2008.0139
  • Callum JL, Waters JH, Shaz BH, Sloan SR, Murphy MF. The AABB recommendations for the choosing wisely campaign of the American Board of Internal Medicine. Transfusion. 2014;54(9):2344–2352. doi:10.1111/trf.12802
  • Aapro M, Chernov VM, Gladkov OA, et al. Practical recommendations for the treatment of anemia in cancer patients. Malignant Tumors. 2016;4:368–377.
  • Bohlius J, Bohlke K, Castelli R, et al. Management of Cancer-Associated anemia with erythropoiesis-stimulating agents: ASCO/ASH Clinical Practice Guideline Update. J Clin Oncol. 2019;37(15):1336–1351. doi:10.1200/JCO.18.02142
  • 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–iv110. doi:10.1093/annonc/mdx758
  • Carson JL, Carless PA, Hebert PC. Transfusion thresholds and other strategies for guiding allogeneic red blood cell transfusion. Cochrane Database Syst Rev. 2018;4(4):CD002042.
  • Goodnough LT. Risks of blood transfusion. Anesthesiol Clin North Am. 2005;23(2):241–252. doi:10.1016/j.atc.2004.07.004
  • Khorana AA, Francis CW, Blumberg N, Culakova E, Refaai MA, Lyman GH. Blood transfusions, thrombosis, and mortality in hospitalized patients with cancer. Arch Intern Med. 2008;168(21):2377–2381. doi:10.1001/archinte.168.21.2377
  • Goubran HA, Elemary M, Radosevich M, Seghatchian J, El-Ekiaby M, Burnouf T. Impact of transfusion on cancer growth and outcome. Cancer Growth Metastasis. 2016;9:1–8. doi:10.4137/CGM.S32797
  • De Oliveira GS Jr, Schink JC, Buoy C, et al. The association between allogeneic perioperative blood transfusion on tumour recurrence and survival in patients with advanced ovarian cancer. Transfus Med. 2012;22(2):97–103. doi:10.1111/j.1365-3148.2011.01122.x
  • Youssef LA, Spitalnik SL. Transfusion-related immunomodulation: a reappraisal. Curr Opin Hematol. 2017;24(6):551–557. doi:10.1097/MOH.0000000000000376
  • Hedenus M, Adriansson M, San Miguel J, et al.; Darbepoetin Alfa 20000161 Study Group. Efficacy and safety of darbepoetin alfa in anaemic patients with lymphoproliferative malignancies: a randomized, double-blind, placebo-controlled study. Br J Haematol. 2003;122(3):394–403. doi:10.1046/j.1365-2141.2003.04448.x
  • Littlewood TJ, Bajetta E, Nortier JW, Vercammen E, Rapoport B; Epoetin Alfa Study Group. 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(11):2865–2874. doi:10.1200/JCO.2001.19.11.2865
  • Vansteenkiste J, Pirker R, Massuti B, et al. Double-blind, placebocontrolled, randomized phase III trial of darbepoetin alfa in lung cancer patients receiving chemotherapy. J Natl Cancer Inst. 2002;94(16):1211–1220. doi:10.1093/jnci/94.16.1211
  • Tonia T, Mettler A, Robert N, et al. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst Rev. 2012;12:CD003407. doi:10.1002/14651858.CD003407.pub5
  • Bohlius J, Tonia T, Nüesch E, et al. Effects of erythropoiesis-stimulating agents on fatigue- and anaemia-related symptoms in cancer patients: systematic review and meta-analyses of published and unpublished data. Br J Cancer. 2014;111(1):33–45. doi:10.1038/bjc.2014.171
  • Aapro M. An update on twenty years of anemia management with erythropoiesis-stimulating agents in nephrology and oncology/hematology. Oncologist. 2009;14(Suppl 1):1–5. doi:10.1634/theoncologist.2009-S1-1
  • Bohlius J, Weingart O, Trelle S, Engert A. Cancer-related anemia and recombinant human erythropoietin-an updated overview. Nat Clin Pract Oncol. 2006;3(3):152–164. doi:10.1038/ncponc0451
  • U.S. Food and Drug Administration. Biosimilar and Interchangeable Products; 2017. Available from: https://www.fda.gov/drugs/biosimilars/biosimilar-and-interchangeable-products. Accessed February 06, 2021.
  • U.S. Food and Drug Administration. Oncologic drugs advisory committee meeting. ‟Epoetin Hospira”, a proposed biosimilar to US-licensed Epogen/Procrit; 2017. Available from: https://www.fda.gov/media/105760/download. Accessed February 06, 2021.
  • US Food and Drug Administration. FDA approves Retacrit as a biosimilar to Epogen/Procrit. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-retacrit-biosimilar-epogenprocrit. Accesed June 18, 2021.
  • Smith RE Jr, Aapro MS, Ludwig H, et al. Darbepoetin alpha for the treatment of anemia in patients with active cancer not receiving chemotherapy or radiotherapy: results of a Phase III, multicenter, randomized, double-blind, placebo-controlled study. J Clin Oncol. 2008;26(7):1040–1050. doi:10.1200/JCO.2007.14.2885
  • Gordon D, Nichols G, Ben‐Jacob A, Tomita D, Lillie T, Miller C. 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(6):715–724. doi:10.1634/theoncologist.2007-0241
  • Grant MD, Piper M, Bohlius J, et al. Epoetin and darbepoetin for managing anemia in patients undergoing cancer treatment: comparative effectiveness update [Internet]. Rockville (MD): Agency for Healthcare Research and Quality (US); 2013. Report No.:13-EHC077-EF.
  • Tonelli M, Hemmelgarn B, Reiman T, et al. Benefits and harms of erythropoiesis-stimulating agents for anemia related to cancer: a meta-analysis. CMAJ. 2009;180(11):E62–71. doi:10.1503/cmaj.090470
  • Bohlius J, Schmidlin K, Brillant C, et al. Recombinant human erythropoiesis stimulating agents and mortality in patients with cancer: a meta-analysis of randomized trials. Lancet. 2009;373(9674):1532–1542. doi:10.1016/S0140-6736(09)60502-X
  • Aapro M, Osterwalder B, Scherhag A, Burger HU. Epoetin-beta treatment in patients with cancer chemotherapy-induced anaemia: the impact of initial haemoglobin and target haemoglobin levels on survival, tumour progression and thromboembolic events. Br J Cancer. 2009;101(12):1961–1971. doi:10.1038/sj.bjc.6605255
  • Bohlius J, Schmidlin K, Brillant C, et al. Erythropoietin or Darbepoetin for patients with cancer: meta-analysis based on individual patient data. Cochrane Database Syst Rev. 2009;2009(3):CD007303.
  • Lyman GH, Bohlke K, Khorana AA, et al.; American Society of Clinical Oncology. Venous thromboembolism prophylaxis and treatment in patients with cancer: american society of clinical oncology clinical practice guideline update 2014. J Clin Oncol. 2015;33(6):654–656. doi:10.1200/JCO.2014.59.7351
  • Donnellan E, Khorana AA. Cancer and venous thromboembolic disease: a review. Oncologist. 2017;22(2):199–207. doi:10.1634/theoncologist.2016-0214
  • Jelkmann W, Bohlius J, Hallek M, Sytkowski AJ. The erythropoietin receptor in normal and cancer tissues. Crit Rev Oncol Hematol. 2008;67(1):39–61.
  • Sinclair AM, Todd MD, Forsythe K, Knox SJ, Elliott S, Begley CG. Expression and function of erythropoietin receptors in tumors: implications for the use of erythropoiesis-stimulating agents in cancer patients. Cancer. 2007;110(3):477–488. doi:10.1002/cncr.22832
  • 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. doi:10.1038/sj.bjc.6605498
  • Engert A, Josting A, Haverkamp H, et al. Epoetin alfa in patients with advanced-stage Hodgkin’s lymphoma: results of the randomized placebo-controlled GHSG HD15EPO trial. J Clin Oncol. 2010;28(13):2239–2245. doi:10.1200/JCO.2009.25.1835
  • Nitz U, Gluz O, Zuna I, et al. West German Study Group. Final results from the prospective phase III WSG-ARA trial: impact of adjuvant darbepoetin alfa on event-free survival in early breast cancer. Ann Oncol. 2014;25(1):75–80. doi:10.1093/annonc/mdt505
  • Pirker R, Ramlau RA, Schuette W, et al. Safety and efficacy of darbepoetin alpha in previously untreated extensive-stage small-cell lung cancer treated with platinum plus etoposide. J Clin Oncol. 2008;26(14):2342–2349. doi:10.1200/JCO.2007.15.0748
  • McKoy JM, Stonecash RE, Cournoyer D, et al. Epoetin-associated pure red cell aplasia: past, present, and future considerations. Transfusion. 2008;48(8):1754–1762. doi:10.1111/j.1537-2995.2008.01749.x
  • Bohlius J, Bohlke K, Castelli R, et al. Management of cancer-associated anemia with erythropoiesis-stimulating agents: ASCO/ASH clinical practice guideline update. Blood Adv. 2019;3(8):1197–1210. doi:10.1182/bloodadvances.2018030387
  • Abdel-Razeq H, Hashem H. Recent update in the pathogenesis and treatment of chemotherapy and cancer induced anemia. Crit Rev Oncol Hematol. 2020;145:102837. doi:10.1016/j.critrevonc.2019.102837
  • Weiss G, Ganz T, Goodnough LT. Anemia of inflammation. Blood. 2019;133(1):40–50. doi:10.1182/blood-2018-06-856500
  • Toblli JE, Angerosa M. Optimizing iron delivery in the management of anemia: patient considerations and the role of ferric carboxymaltose. Drug Des Dev Ther. 2014;8:2475–2491. doi:10.2147/DDDT.S55499
  • Baribeault D, Auerbach M. Iron replacement therapy in cancer-related anemia. Am J Health Syst Pharm. 2011;68(10 Suppl 1):S4–S14. doi:10.2146/ajhp110039
  • Mhaskar R, Wao H, Miladinovic B, Kumar A, Djulbegovic B. The role of iron in the management of chemotherapy-induced anemia in cancer patients receiving erythropoiesis stimulating agents. Cochrane Database Syst Rev. 2016;2:CD009624. doi:10.1002/14651858.CD009624.pub2
  • Auerbach M, Silberstein PT, Webb RT, et al. Darbepoetin alfa 300 or 500 mg once every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. Am J Hematol. 2010;85(9):655–663. doi:10.1002/ajh.21779
  • Bastit L, Vandebroek A, 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. doi:10.1200/JCO.2006.10.4620
  • Gafter-Gvili A, Rozen-Zvi B, Vidal L, et al. Intravenous iron supplementation for the treatment of chemotherapy anaemia: systematic review and meta-analysis of randomized controlled studies. Acta Oncol. 2013;52(1):18–29. doi:10.3109/0284186X.2012.702921
  • Steensma DP, Sloan JA, Dakhil SR, et al. Phase III, randomized study of the effects of parenteral iron, oral iron, or no iron supplementation on the erythropoietic response to darbepoetin alfa for patients with chemotherapy-associated anemia. J Clin Oncol. 2011;29(1):97–105. doi:10.1200/JCO.2010.30.3644
  • Jang JH, 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. doi:10.1371/journal.pmed.1003091
  • Steensma DP, Sasu BJ, Sloan JA, Tomita DK, Loprinzi CL. Serum hepcidin levels predict response to intravenous iron and darbepoetin in chemotherapy-associated anemia. Blood. 2015;125(23):3669–3671. doi:10.1182/blood-2015-03-636407
  • Gozzelino R, Arosio P. Iron homeostasis in health and disease. Int J Mol Sci. 2016;17(1):130. doi:10.3390/ijms17010130
  • González-Chávez SA, Arévalo-Gallegos S, Rascón-Cruz Q. Lactoferrin: structure, function and applications. Int J Antimicrob Agents. 2009;33(4):301.e1–8. doi:10.1016/j.ijantimicag.2008.07.020
  • Macciò A, Madeddu C, Gramignano G, Mulas C, Sanna E, Mantovani G. Efficacy and safety of oral lactoferrin supplementation in combination with rHuEPO-b for the treatment of anemia in advanced cancer patients undergoing chemotherapy: open-label, randomized controlled study. Oncologist. 2010;15(8):894–902. doi:10.1634/theoncologist.2010-0020
  • Cooke KS, Hinkle B, Salimi-Moosavi H, et al. A fully human anti-hepcidin antibody modulates iron metabolism in both mice and nonhuman primates. Blood. 2013;122(17):3054–3061. doi:10.1182/blood-2013-06-505792
  • 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. doi:10.1186/s13045-017-0427-x
  • Park EJ, Choi J, Lee KC, Na DH. Emerging PEGylated non-biologic drugs. Expert Opin Emerg Drugs. 2019;24(2):107–119. doi:10.1080/14728214.2019.1604684
  • Georgiev P, Lazarolu M, Ocroteala L, et al. The anti-hepcidin Spiegelmer Lexapeptid Pegol (NOX-H94) as treatment of anemia of chronic disease in patients with multiple myeloma, low grade lymphoma and CLL: a phase II pilot study. AACR Meeting; 2014:Abstract 3847.
  • Haase VH. Therapeutic targeting of the HIF oxygen-sensing pathway: lessons learned from clinical studies. Exp Cell Res. 2017;356(2):160–165. doi:10.1016/j.yexcr.2017.05.004
  • Forristal CE, Levesque JP. Targeting the hypoxia-sensing pathway in clinical hematology. Stem Cell Transl Med. 2014;3(2):135–140. doi:10.5966/sctm.2013-0134
  • Wan J, Chai H, Yu Z, et al. HIF-1α effects on angiogenic potential in human small cell lung carcinoma. J Exp Clin Cancer Res. 2011;30(1):77. doi:10.1186/1756-9966-30-77
  • Seeley TW, Sternlicht MD, Klaus SJ, Neff TB, Liu DY. Induction of erythropoiesis by hypoxia-inducible factor prolyl hydroxylase inhibitors without promotion of tumor initiation, progression, or metastasis in a VEGF-sensitive model of spontaneous breast cancer. Hypoxia (Auckl). 2017;5:1–9. doi:10.2147/HP.S130526
  • Bayliss TJ, Smith JT, Schuster M, Dragnev KH, Rigas JR. A humanized anti-IL-6 antibody (ALD518) in non-small cell lung cancer. Expert Opin Biol Ther. 2011;11(12):1663–1668. doi:10.1517/14712598.2011.627850
  • Coward J, Kulbe H, Chakravarty P, et al. Interleukin-6 as a therapeutic target in human ovarian cancer. Clin Cancer Res. 2011;17(18):6083–6096. doi:10.1158/1078-0432.CCR-11-0945
  • Pauklin S, Vallier L. Activin/nodal signaling in stem cells. Development. 2015;142(4):607–619. doi:10.1242/dev.091769
  • Miller KL, Carlino JA, Ogawa Y, Avis PD, Carroll KG. Alterations in erythropoiesis in TGF-b 1-treated mice. Exp Hematol. 1992;20(8):951–956.
  • Kang YJ, Shin JW, Yoon JH, et al. Inhibition of erythropoiesis by Smad6 in human cord blood hematopoietic stem cells. Biochem Biophys Res Commun. 2011;423(4):750–756. doi:10.1016/j.bbrc.2012.06.031
  • Brancaleoni V, Nava I, Delbini P, Duca L, Motta I. Activin receptor-ligand trap for the treatment of β-thalassemia: a serendipitous discovery. Mediterr J Hematol Infect Dis. 2020;12(1):e2020075. doi:10.4084/mjhid.2020.075
  • Testa U, Castelli G, Elvira P. Experimental and investigational therapies for chemotherapy-induced anemia. Expert Opin Investig Drugs. 2015;24(11):1433–1445. doi:10.1517/13543784.2015.1085505
  • Patel B, Moosavi L. Luspatercept. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing LLC; July 10, 2020.
  • Fenaux P, Kiladjian JJ, Platzbecker U. Luspatercept for the treatment of anemia in myelodysplastic syndromes and primary myelofibrosis. Blood. 2019;133(8):790–794. doi:10.1182/blood-2018-11-876888
  • Fenaux P, Platzbecker U, Mufti GJ, et al. Luspatercept in patients with lower-risk myelodysplastic syndromes. N Engl J Med. 2020;382(2):140–151. doi:10.1056/NEJMoa1908892
  • Kubasch AS, Fenaux P, Platzbecker U. Development of luspatercept to treat ineffective erythropoiesis. Blood Adv. 2021;5(5):1565–1575. doi:10.1182/bloodadvances.2020002177
  • Feld J, Navada SC, Silverman LR. Myelo-deception: luspatercept & TGF-Beta ligand traps in myeloid diseases & anemia. Leuk Res. 2020;97:106430. doi:10.1016/j.leukres.2020.106430
  • Raftopoulos H, Laadem A, Hesketh PJ, et al. Sotatercept (ACE-011) for the treatment of chemotherapy-induced anemia in patients with metastatic breast cancer or advanced or metastatic solid tumors treated with platinum-based chemotherapeutic regimens: results from two Phase 2 studies. Support Care Cancer. 2016;24(4):1517–1525. doi:10.1007/s00520-015-2929-9
  • Layer G, Reichelt J, Jahn D, Heinz DW. Structure and function of enzymes in heme biosynthesis. Protein Sci. 2010;19(6):1137–1161. doi:10.1002/pro.405
  • Madeddu C, Mantovani G, Gramignano G, Astara G, Macciò A. Muscle wasting as main evidence of energy impairment in cancer cachexia: future therapeutic approaches. Future Oncol. 2015;11(19):2697–2710. doi:10.2217/fon.15.195
  • Friesen DE, Baracos VE, Tuszynski JA. Modeling the energetic cost of cancer as a result of altered energy metabolism: implications for cachexia. Theor Biol Med Model. 2015;12:17. doi:10.1186/s12976-015-0015-0
  • Santino A, Scarano A, De Santis S, De Benedictis M, Giovinazzo G, Chieppa M. Gut microbiota modulation and anti-inflammatory properties of dietary polyphenols in IBD: new and consolidated perspectives. Curr Pharm Des. 2017;23(16):2344–2351. doi:10.2174/1381612823666170207145420
  • Rahman I, Biswas SK, Kirkham PA. Regulation of inflammation and redox signaling by dietary polyphenols. Biochem Pharmacol. 2006;72(11):1439–1452. doi:10.1016/j.bcp.2006.07.004
  • Yahfoufi N, Alsadi N, Jambi M, Matar C. The Immunomodulatory and anti-inflammatory role of polyphenols. Nutrients. 2018;10(11):1618. doi:10.3390/nu10111618
  • Fatih N, Camberlein E, Island ML, et al. Natural and synthetic STAT3 inhibitors reduce hepcidin expression in differentiated mouse hepatocytes expressing the active phosphorylated STAT3 form. J Mol Med (Berl). 2010;88(5):477–486. doi:10.1007/s00109-009-0588-3
  • Lainé F, Laviolle B, Bardou-Jacquet E, et al. Curcuma decreases serum hepcidin levels in healthy volunteers: a placebo-controlled, randomized, double-blind, cross-over study. Fundam Clin Pharm. 2017;31(5):567–573. doi:10.1111/fcp.12288
  • Jiao Y, Wilkinson J, Di X, et al. Curcumin, a cancer chemopreventive and chemotherapeutic agent, is a biologically active iron chelator. Blood. 2009;113(2):462–469. doi:10.1182/blood-2008-05-155952
  • Mohammadi E, Tamaddoni A, Qujeq D, et al. An investigation of the effects of curcumin on iron overload, hepcidin level, and liver function in β-thalassemia major patients: a double-blind randomized controlled clinical trial. Phytother Res. 2018;32(9):1828–1835. doi:10.1002/ptr.6118
  • Macciò A, Gramignano G, Madeddu C. A multitargeted treatment approach for anemia and cachexia in metastatic castration-resistant prostate cancer. J Pain Symptom Manage. 2015;50(2):e1–e4. doi:10.1016/j.jpainsymman.2015.04.014
  • Macciò A, Gramignano G, Madeddu C. Surprising results of a supportive integrated therapy in myelofibrosis. Nutrition. 2015;31(1):239–243. doi:10.1016/j.nut.2014.07.016
  • Endo K, Tsuji A, Kondo S, Wakisaka N, Murono S, Yoshizaki T. Carnitine is associated with fatigue following chemoradiotherapy for head and neck cancer. Acta Otolaryngol. 2015;135(8):846–852. doi:10.3109/00016489.2015.1030769
  • Hockenberry MJ, Hooke MC, Gregurich M, McCarthy K. Carnitine plasma levels and fatigue in children/adolescents receiving cisplatin, ifosfamide, or doxorubicin. J Pediatr Hematol Oncol. 2009;31(9):664–669. doi:10.1097/MPH.0b013e3181b259a7
  • Cruciani RA, Dvorkin E, Homel P, et al. L-carnitine supplementation in patients with advanced cancer and carnitine deficiency: a double-blind, placebo-controlled study. J Pain Symptom Manage. 2009;37(4):622–631. doi:10.1016/j.jpainsymman.2008.03.021
  • Morris MS, Jacques PF, Rosenberg IH, Selhub J. Folate and vitamin B-12 status in relation to anemia, macrocytosis, and cognitive impairment in older Americans in the age of folic acid fortification. Am J Clin Nutr. 2007;85(1):193–200. doi:10.1093/ajcn/85.1.193
  • Lonsdale D. A review of the biochemistry, metabolism and clinical benefits of thiamin(e) and its derivatives. Evid Based Complement Alternat Med. 2006;3(1):49–59. doi:10.1093/ecam/nek009
  • Parra M, Stahl S, Hellmann H. Vitamin B₆ and its role in cell metabolism and physiology. Cells. 2018;7(7):84. doi:10.3390/cells7070084
  • Smith EM, Tangpricha V. Vitamin D and anemia: insights into an emerging association. Curr Opin Endocrinol Diabetes Obes. 2015;22(6):432–438. doi:10.1097/MED.0000000000000199
  • Zughaier SM, Alvarez JA, Sloan JH, Konrad RJ, Tangpricha V. The role of vitamin D in regulating the iron-hepcidin-ferroportin axis in monocytes. J Clin Transl Endocrinol. 2014;1(1):19–25. doi:10.1016/j.jcte.2014.01.003
  • Perlstein TS, Pande R, Berliner N, Vanasse GJ. Prevalence of 25-hydroxyvitamin D deficiency in subgroups of elderly persons with anemia: association with anemia of inflammation. Blood. 2011;117(10):2800–2806. doi:10.1182/blood-2010-09-309708
  • Icardi A, Paoletti E, De Nicola L, Mazzaferro S, Russo R, Cozzolino M. Renal anaemia and EPO hyporesponsiveness associated with vitamin D deficiency: the potential role of inflammation. Nephrol Dial Transplant. 2013;28(7):1672–1679. doi:10.1093/ndt/gft021
  • Smith EM, Alvarez JA, Kearns MD, et al. High-dose vitamin D3 reduces circulating hepcidin concentrations: a pilot, randomized, double-blind, placebo-controlled trial in healthy adults. Clin Nutr. 2017;36(4):980–985. doi:10.1016/j.clnu.2016.06.015
  • Moran-Lev H, Galai T, Yerushalmy-Feler A, et al. Vitamin D decreases hepcidin and inflammatory markers in newly diagnosed inflammatory bowel disease paediatric patients: a Prospective Study. J Crohn’s Colitis. 2019;13(10):1287–1291. doi:10.1093/ecco-jcc/jjz056
  • Pasini E, Corsetti G, Romano C, et al. Management of anaemia of chronic disease: beyond iron-only supplementation. Nutrients. 2021;13(1):237. doi:10.3390/nu13010237
  • Hallberg L, Brune M, Rossander L. Iron absorption in man: ascorbic acid and dose-dependent inhibition by phytate. Am J Clin Nutr. 1989;49(1):140–144. doi:10.1093/ajcn/49.1.140
  • Lane DJ, Richardson DR. The active role of vitamin C in mammalian iron metabolism: much more than just enhanced iron absorption! Free Radic Biol Med. 2014;75:69–83. doi:10.1016/j.freeradbiomed.2014.07.007
  • Chiu PF, Ko SY, Chang CC. Vitamin C affects the expression of hepcidin and erythropoietin receptor in HepG2 cells. J Ren Nutr. 2012;22(3):373–376. doi:10.1053/j.jrn.2011.09.007
  • Attallah N, Osman-Malik Y, Frinak S, Besarab A. Effect of intravenous ascorbic acid in hemodialysis patients with EPO-hyporesponsive anemia and hyperferritinemia. Am J Kidney Dis. 2006;47(4):644–654. doi:10.1053/j.ajkd.2005.12.025
  • Barni S, Gascòn P, Petrelli F, et al. Position paper on management of iron deficiency in adult cancer patients. Expert Rev Hematol. 2017;10(8):685–695. doi:10.1080/17474086.2017.1343140
  • Steinmetz T, Tschechne B, Harlin O, et al. Clinical experience with ferric carboxymaltose in the treatment of cancer- and chemotherapy-associated anaemia. Ann Oncol. 2013;24(2):475–482. doi:10.1093/annonc/mds338
  • Toledano A, Luporsi E, Morere JF, et al. Clinical use of ferric carboxymaltose in patients with solid tumours or haematological malignancies in France. Support Care Cancer. 2016;24(1):67–75. doi:10.1007/s00520-015-2728-3