Intravenous Iron Therapy to Treat Anemia in Oncology: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Jayne Lim.

Anemia is a common problem when patients present with cancer, and it can worsen during treatment. Anemia can directly impact the cognitive and physical quality of life and may impair fitness for oncological therapy. The most common cause of anemia is iron deficiency. Newer intravenous (IV) iron formulations offer a safe and rapidly effective treatment option. 

  • anemia
  • oncology
  • mapping
  • randomized controlled trials

1. Introduction

Anemia is a common problem in cancer patients; one-third have anemia at diagnosis, and half develop anemia during chemotherapy and/or radiation therapy [1]. Anemia is defined by the World Health Organization as a hemoglobin concentration (Hb) < 120 g/L in women and <130 g/L in men [1]. Anemia is caused by one or more primary mechanisms: blood loss, hemolysis, and reduction in erythropoiesis [2]. Blood loss is a direct result of gastrointestinal tumors and bone marrow replacement by cancer cells. Bone marrow suppression, cancer-related cachexia, and hepcidin-mediated iron sequestration are indirect effects of inflammation [2]. Anemia is associated with fatigue, impaired physical function, and decreased quality of life [3]. The severity of anemia is associated with a decreased quality of life [4]. The impact of anemia on “fitness” has a direct impact on oncological therapy, where performance status impacts fitness for treatment [5]. This is particularly relevant prior to surgery when either neoadjuvant or adjuvant therapy is indicated [6].
The problem of anemia has been highlighted in surgical patients, where one in three has preoperative anemia [7]. Preoperative anemia itself is independently associated with increased morbidity and mortality, as well as the risk of red blood cell (RBC) transfusion, postoperative complications, and increased hospital stay [8]. Blood loss during operation and hospital stay results in most patients being discharged with anemia, one in four with Hb < 100 g/L, which is also associated with increased rates of readmission for complications and delayed recovery [9]. After major surgery for solid tumors, which often require adjuvant therapy, a delay in initiation is associated with inferior overall treatment and survival outcomes in several meta-analyses [10,11][10][11].
Current treatment options for anemia include the use of blood transfusions, erythroid-stimulating agents (ESAs), or iron [12,13][12][13]. Although the use of blood transfusions can rapidly increase Hb, there is caution as database analyses suggest that it may be associated with adverse outcomes and potentially an immunomodulated risk of increased cancer recurrence. ESAs are effective in increasing Hb, and guidelines limit administration to Hb levels less than 100 g/L due to some safety concerns [14,15,16,17][14][15][16][17]. Prospective studies and meta-analyses investigating ESAs that have followed did not demonstrate negative effects [18,19,20][18][19][20]. Iron therapy has traditionally focused on oral iron. However, side effects in the gastrointestinal tract often mean that only a minority of cancer patients, who may already experience nausea, diarrhea, or constipation as a side effect of their cancer therapy, can tolerate oral iron. Intravenous (IV) iron has had limited use due to historical concerns about anaphylaxis (high molecular weight iron dextran products are no longer available for clinical use) or low doses (200 mg of iron sucrose) [21].

2. Surgery

There were five RCTs performed on colorectal cancer patients undergoing elective surgery; four evaluated the efficacy of preoperative IV iron, and one compared different formulations of IV iron in the postoperative setting [35,36,41,43,49][22][23][24][25][26]. The studies included patients with anemia (defined as Hb <100 g/L–≤135 g/L), with two studies including iron parameters. In general, the most common primary endpoint was a change in Hb and a decrease in transfusion rates. IV iron increased Hb levels before and after surgery (7.8 g/L), but secondary endpoints of quality of life scores, length of hospital stay, postoperative complications, and adverse events showed limited differences between groups. In summary, the use of IV iron appears to correct laboratory values of Hb and may reduce the need for blood transfusion, but clinical trials have not shown direct patient benefit on clinical outcomes.

Detailed Description of the IVICA and FIT Trials

IVICA included 116 patients with anemia defined as 10 g/L below WHO definition, scheduled for elective colorectal cancer surgery a minimum of 14 days before surgery [41][24]. The intervention of 1000 mg to 2000 mg of IV ferric carboxymaltose (FCM) based on the patient’s Hb and weight was compared to 200 mg of oral ferrous sulfate twice daily until the day of surgery. There was no difference in the primary endpoint of blood transfusion between the groups. Hb levels were significantly higher in the IV iron group at the time of surgery, with a greater proportion in the IV iron group achieving Hb normalization on the day of surgery compared to oral iron (90% vs. 75%; p = 0.048). Secondary endpoints did not show differences in the rate of complications (including infection) or postoperative length of hospital stay. A secondary manuscript suggested that compared to oral iron, IV iron improved quality of life both on the day of surgery and on the first outpatient visit (2–3 months after hospital discharge) [50][27]. FIT enrolled 202 patients with anemia defined as WHO definitions and iron deficiency defined as transferrin saturation (TSAT) < 20% who underwent curative resection for colorectal cancer [49][26]. Patients were divided into two groups receiving either 1000 mg to 2000 mg IV FCM in a single dose within four weeks of surgery or 200 mg of oral ferrous fumarate three times daily until the day before surgery and followed for 6 months. Oral iron was continued if anemia persisted after surgery. The primary endpoint reported that IV iron was not superior to oral iron in normalizing Hb levels on the day of surgery. However, IV iron led to a significant increase in Hb normalization at 1 month with 60% participants compared with 21% with oral iron (RR 2.92; 95% CI: 1.87–4.58; p < 0.0001), 2 months with 76% IV iron compared with 45% oral iron (RR 1.69; 95% CI: 1.29–2.23; p < 0.0001), and 3 months with 76% IV iron compared with 43% with oral iron (RR 1.76; 95% CI: 1.34–2.32; p < 0.0001). No differences were observed in RBC transfusions, postoperative complications, length of hospital stay, and mortality from baseline before surgery and postoperatively. Overall, FIT observed that IV iron increases Hb preoperatively, with the optimal benefits seen 1–3 months after surgery. These results paint an encouraging picture, with 33% of patients receiving adjuvant chemotherapy. IV iron was associated with sustained benefit three months after single-dose treatment.

3. Adjuvant Therapy after Surgery

One RCT evaluated 44 anemic women with Hb < 109 g/L and gynecological cancer who received platinum-based adjuvant chemotherapy after previous resection [32][28]. All patients received RBC transfusion prior to chemotherapy in accordance with the standard hospital protocol. Patients were randomly assigned to receive 200 mg of IV iron sucrose (IS) in a single dose or 200 mg of oral ferrous sulfate three times daily and followed until the next cycle of chemotherapy. The results revealed a lower requirement for RBC transfusions in the IV iron group vs. the oral iron group (22.7% vs. 63.6%, respectively, p < 0.01) in the next chemotherapy cycle. Significantly higher Hb and hematocrit were reported in the IV iron group. There was no change in total quality of life scores before and after treatment in both groups.

4. Adjuvant Therapy with ESAs

Ten RCTs evaluated the effects of adding IV iron to ESAs in cancer patients with anemia related to cancer and/or chemotherapy [28,29,30,31,38,39,40,44,47,48][29][30][31][32][33][34][35][36][37][38]. A variety of cancers, including solid tumors (breast cancer, colorectal, lung, and gynecological) and lymphomas, were evaluated. Most studies included anemia defined as Hb < 110 g/L, and seven studies included serum ferritin and TSAT parameters, although the definitions are heterogeneous. Overall, the increase in Hb was the primary endpoint. Secondary endpoints were transfusion rate and quality of life scores. The use of IV iron was associated with significantly increased Hb levels, defined as ≥20 g/L increase from baseline and/or decreased requirement for RBC transfusion (20%) compared to oral iron and ESAs alone.

Detailed Description of Specific Trials

Auerbach et al. were the first to explore the addition of iron therapy to ESAs in the oncology setting [29][30]. One hundred and fifty-seven patients were randomized to four groups receiving either no iron, 325 mg of oral ferrous sulfate twice daily, 100 mg of IV iron dextran bolus injection at each visit (to the calculated dose of iron replacement), or IV iron dextran (total dose infusion calculated with a formula), and followed up for 6 weeks. All patients received 40,000 U of ESAs once weekly. All treatment groups showed a significant increase in Hb from baseline to 6 weeks. The mean increase was 9 g/L, 15 g/L, 25 g/L, and 24 g/L. The two IV iron groups showed a three-fold increase in mean change in Hb. There was a significant improvement in quality of life scores in both IV iron groups. Henry et al. enrolled 187 patients undergoing chemotherapy and randomized in a 1:1:1 ratio to receive either 125 mg of IV sodium ferric gluconate complex (FG) weekly, 325 mg of oral ferrous sulfate three times daily, or no iron for 8 weeks [40][35]. All groups were scheduled to receive 40,000 U of ESAs once weekly. The primary endpoint was met with an increase in Hb without transfusions at 10 weeks. A total of 73% of the IV iron group, 46% of the oral iron group, and 41% with no iron group had a hematopoietic response defined as Hb response (increase > 20 g/L). Bastit et al. assessed 396 patients with non-myeloid malignancies undergoing chemotherapy and Hb < 110 g/L to receive either 200 mg of IV iron + ESAs every three weeks or ESAs every three weeks and standard practice for 16 weeks [31][32]. A higher proportion achieved the Hb target in the IV iron group (86% vs. 73%), defined as Hb ≥ 120 g/L or Hb increase of ≥20 g/L from baseline. Treatment was tolerated in both groups, with no differences in serious adverse events being observed. Auerbach et al. further conducted a double-blind, 2 × 2 factorial RCT in which 243 oncology patients with anemia were randomized to one of four groups (300 µg or 500 µg of ESAs once every three weeks) and (400 μg of IV iron or no iron every three weeks) and followed up for 15 weeks [30][31]. At the end of the intervention period, the average change in Hb was greater in the IV iron groups, and the proportion of patients who achieved Hb was higher in the IV iron groups compared to those without iron. IV iron was associated with improved quality of life scores. The median time to clinical improvement was shorter in the IV iron group than in the no IV iron group (7 weeks vs. 10 weeks). Anthony et al. evaluated 375 oncology patients with Hb ≤ 100 g/L [28][29]. Patients were randomized to receive either IV iron sucrose 3 times per week with ESAs or ESAs only and followed up for 12 weeks. At the end of the intervention, Hb and QoL were significantly higher in the IV iron group compared to the no iron group. Steensma et al. enrolled 502 patients with non-myeloid malignancies undergoing chemotherapy with Hb < 110 g/L to receive either IV FG every 3 weeks, oral iron, or oral placebo at a 1:1:1 ratio for 16 weeks [48][38]. The primary endpoint was the proportion of those who achieved a Hb response ≥ 20 g/L from baseline or Hb < 120 g/L in the absence of transfusions during the preceding 4 weeks. The investigators reported no difference in Hb response, transfusion rate, or quality of life scores in the intention-to-treat cohort. Subsequent per-protocol analysis evaluating those who received at least four of five doses of planned FG doses reported that IV iron was more efficacious in increasing Hb response (80% vs. 67% vs. 65%) and decreasing transfusion rates (9% vs. 13% vs. 13%) [51][39].

5. Adjuvant Therapy without ESAs

Four RCTs compared the safety and efficacy of IV iron monotherapy to treat chemotherapy-induced anemia [27,39,45,46][34][40][41][42]. Patients with lymphoid, colon, solid, and non-myeloid cancers were included. Most studies defined anemia below WHO classification, and Hb increase was the primary endpoint. Hedenus et al. were the first to investigate whether IV iron would increase Hb in patients with lymphoid malignancies and functional iron deficiency (Hb 85–105 g/L and SF > 30 μg/L for women or >40 μg/L for men, TSAT ≤ 20%). Seventeen were randomized to receive IV iron or no treatment (standard care) and followed up at 4, 6, and 8 weeks. The study reported that IV iron significantly increased Hb at the 8-week primary endpoint. Median Hb increase was 21 g/L (range 2–35 g/L) in the IV iron group vs. 9 g/L (range 3–22 g/L). No treatment-related adverse events were reported. AnsariNejad et al. evaluated patients with stage III/IV colon cancer undergoing FOLFOX chemotherapy with Hb ≤ 120 g/L for women and ≤130 g/L for men, SF < 30 µg/L. Sixty patients were randomly assigned to receive single-dose IV iron or oral iron for 8 weeks. There was a significant increase in Hb at the primary endpoint, defined as 6 weeks after treatment with IV iron and 8 weeks of oral iron treatment (138.6 ± 7.4 g/L vs. 116.7 ± 12.8 g/L). Noronha et al. enrolled 148 patients undergoing chemotherapy with Hb < 120 g/L and at least one characteristic ID: SF < 100 µg/L, TSAT < 20%, or hypochromic RBC > 10% were enrolled. Patients were randomly assigned to receive IV iron or oral iron. The study found no difference in the mean increase in Hb at 6 weeks, and transfusion rate and quality of life scores were similar between the two arms. More recently, Makharadze et al. enrolled 244 patients who underwent chemotherapy with Hb 80–110 g/L to receive either IV iron or placebo and follow up for 18 weeks. The primary endpoint was defined as a decrease in Hb = 5 g/L from baseline at weeks 3 to 18. IV iron was associated with a maintained Hb (50.8% vs. 35.3%; p = 0.01). IV iron was well tolerated.

6. Radiotherapy

Two RCTs evaluated IV iron in patients with cervical cancer who underwent chemoradiotherapy or primary radiotherapy [34,42][43][44]. Kim et al. evaluated 75 women with Hb ≤ 120 g/L and locally advanced cervical cancer undergoing concurrent chemoradiotherapy. Patients were randomized to receive either IV iron or no iron at the start of each cycle of treatment. IV iron was administered when Hb levels = 100–120 g/L. The results revealed a significant reduction in transfusion rates and units in the IV iron arm over a six-cycle treatment period and increased Hb levels. Dreyer et al. enrolled 43 patients scheduled for primary radiotherapy with Hb ≤ 120 g/L and randomized to receive either IV iron + limited transfusion or standard transfusion. In the IV iron and limited transfusion group, a steady rise in Hb over 12 weeks was observed, while the transfusion and no iron group showed a 5 g/L decline in Hb after treatment.

7. Palliative

A recent study explored the feasibility of performing a large definitive RCT in patients with advanced solid tumors [33][45]. Study authors Dickson et al. conducted a double-blind, placebo-controlled trial in which 34 patients with anemia defined by WHO definitions with fatigue and performance status <2 were randomized in a 1:1 ratio to receive either IV iron or placebo (normal saline) and followed up for 8 weeks. The trial was feasible according to the recruitment rate (47%) and study attrition (26%). Hb increased in the IV iron group at 4 and 8 weeks. Hb levels increased in the IV group (39% vs. 8%) compared to the placebo group at 8 weeks.

References

  1. 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, 444–454.
  2. Gilreath, J.A.; Rodgers, G.M. How I treat cancer-associated anemia. Blood 2020, 136, 801–813.
  3. Cella, D.; Kallich, J.; McDermott, A.; Xu, X. The longitudinal relationship of hemoglobin, fatigue and quality of life in anemic cancer patients: Results from five randomized clinical trials. Ann. Oncol. 2004, 15, 979–986.
  4. Lind, M.; Vernon, C.; Cruickshank, D.; Wilkinson, P.; Littlewood, T.; Stuart, N.; Jenkinson, C.; Grey-Amante, P.; Doll, H.; Wild, D. The level of haemoglobin in anaemic cancer patients correlates positively with quality of life. Br. J. Cancer 2002, 86, 1243–1249.
  5. West, H.J.; Jin, J.O. JAMA Oncology Patient Page. Performance Status in Patients With Cancer. JAMA Oncol. 2015, 1, 998.
  6. Nachiappan, S.; Askari, A.; Mamidanna, R.; Munasinghe, A.; Currie, A.; Stebbing, J.; Faiz, O. The impact of adjuvant chemotherapy timing on overall survival following colorectal cancer resection. Eur. J. Surg. Oncol. 2015, 41, 1636–1644.
  7. Shander, A.; Knight, K.; Thurer, R.; Adamson, J.; Spence, R. Prevalence and outcomes of anemia in surgery: A systematic review of the literature. Am. J. Med. 2004, 116 (Suppl. S7A), 58S–69S.
  8. Munoz, M.; Gomez-Ramirez, S.; Campos, A.; Ruiz, J.; Liumbruno, G.M. Pre-operative anaemia: Prevalence, consequences and approaches to management. Blood Transfus. 2015, 13, 370–379.
  9. Collaborative, P.O.S. The management of peri-operative anaemia in patients undergoing major abdominal surgery in Australia and New Zealand: A prospective cohort study. Med. J. Aust. 2022, 217, 487–493.
  10. Raphael, M.J.; Biagi, J.J.; Kong, W.; Mates, M.; Booth, C.M.; Mackillop, W.J. The relationship between time to initiation of adjuvant chemotherapy and survival in breast cancer: A systematic review and meta-analysis. Breast Cancer Res. Treat. 2016, 160, 17–28.
  11. Biagi, J.J.; Raphael, M.J.; Mackillop, W.J.; Kong, W.; King, W.D.; Booth, C.M. Association between time to initiation of adjuvant chemotherapy and survival in colorectal cancer: A systematic review and meta-analysis. JAMA 2011, 305, 2335–2342.
  12. Rodgers, G.M. A perspective on the evolution of management of cancer- and chemotherapy-induced anemia. J. Natl. Compr. Canc. Netw. 2012, 10, 434–437.
  13. Aapro, M.; Beguin, Y.; Bokemeyer, C.; Dicato, M.; Gascón, P.; Glaspy, J.; Hofmann, A.; Link, H.; Littlewood, T.; Ludwig, H.; et al. Management of anaemia and iron deficiency in patients with cancer: ESMO Clinical Practice Guidelines. Ann. Oncol. 2018, 29, iv96–iv110.
  14. Henke, M.; Laszig, R.; Rube, C.; Schafer, U.; Haase, K.D.; Schilcher, B.; Mose, S.; Beer, K.T.; Burger, U.; Dougherty, 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.
  15. Bokemeyer, C.; Aapro, M.S.; Courdi, A.; Foubert, J.; Link, H.; Osterborg, A.; Repetto, L.; Soubeyran, P. EORTC guidelines for the use of erythropoietic proteins in anaemic patients with cancer. Eur. J. Cancer 2004, 40, 2201–2216.
  16. Leyland-Jones, B.; Investigators, B.; Study, G. Breast cancer trial with erythropoietin terminated unexpectedly. Lancet Oncol. 2003, 4, 459–460.
  17. Bohlius, J.; Langensiepen, S.; Schwarzer, G.; Seidenfeld, J.; Piper, M.; Bennet, C.; Engert, A. Erythropoietin for patients with malignant disease. Cochrane Database Syst. Rev. 2004, CD003407.
  18. Gascon, P.; Nagarkar, R.; Smakal, M.; Syrigos, K.N.; Barrios, C.H.; Sanchez, J.C.; Zhang, L.; Henry, D.H.; Gordon, D.; Hirsh, V.; 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, 190–202.
  19. Glaspy, J.; Crawford, J.; Vansteenkiste, J.; Henry, D.; Rao, S.; Bowers, P.; Berlin, J.A.; Tomita, D.; Bridges, K.; Ludwig, H. Erythropoiesis-stimulating agents in oncology: A study-level meta-analysis of survival and other safety outcomes. Br. J. Cancer 2010, 102, 301–315.
  20. Vansteenkiste, J.; Glaspy, J.; Henry, D.; Ludwig, H.; Pirker, R.; Tomita, D.; Collins, H.; Crawford, J. Benefits and risks of using erythropoiesis-stimulating agents (ESAs) in lung cancer patients: Study-level and patient-level meta-analyses. Lung Cancer 2012, 76, 478–485.
  21. Avni, T.; Bieber, A.; Grossman, A.; Green, H.; Leibovici, L.; Gafter-Gvili, A. The safety of intravenous iron preparations: Systematic review and meta-analysis. Mayo Clin. Proc. 2015, 90, 12–23.
  22. Edwards, T.J.; Noble, E.J.; Durran, A.; Mellor, N.; Hosie, K.B. Randomized clinical trial of preoperative intravenous iron sucrose to reduce blood transfusion in anaemic patients after colorectal cancer surgery. Br. J. Surg. 2009, 96, 1122–1128.
  23. Fung, P.L.P.; Lau, V.N.M.; Ng, F.F.; Leung, W.W.; Mak, T.W.C.; Lee, A. Perioperative changes in haemoglobin and ferritin concentrations from preoperative intravenous iron isomaltoside for iron deficiency anaemia in patients with colorectal cancer: A pilot randomised controlled trial. PLoS ONE 2022, 17, e0270640.
  24. Keeler, B.D.; Simpson, J.A.; Ng, O.; Padmanabhan, H.; Brookes, M.J.; Acheson, A.G.; Group, I.T. Randomized clinical trial of preoperative oral versus intravenous iron in anaemic patients with colorectal cancer. Br. J. Surg. 2017, 104, 214–221.
  25. Laso-Morales, M.J.; Vives, R.; Bisbe, E.; Garcia-Erce, J.A.; Munoz, M.; Martinez-Lopez, F.; Carol-Boeris, F.; Pontes-Garcia, C. Single-dose intravenous ferric carboxymaltose infusion versus multiple fractionated doses of intravenous iron sucrose in the treatment of post-operative anaemia in colorectal cancer patients: A randomised controlled trial. Blood Transfus. 2022, 20, 310–318.
  26. Talboom, K.; Borstlap, W.A.A.; Roodbeen, S.X.; Bruns, E.R.J.; Buskens, C.J.; Hompes, R.; Tytgat, K.; Tuynman, J.B.; Consten, E.C.J.; Heuff, G.; et al. Ferric carboxymaltose infusion versus oral iron supplementation for preoperative iron deficiency anaemia in patients with colorectal cancer (FIT): A multicentre, open-label, randomised, controlled trial. Lancet Haematol. 2023, 10, e250–e260.
  27. Keeler, B.D.; Dickson, E.A.; Simpson, J.A.; Ng, O.; Padmanabhan, H.; Brookes, M.J.; Acheson, A.G.; Group, I.T. The impact of pre-operative intravenous iron on quality of life after colorectal cancer surgery: Outcomes from the intravenous iron in colorectal cancer-associated anaemia (IVICA) trial. Anaesthesia 2019, 74, 714–725.
  28. Dangsuwan, P.; Manchana, T. Blood transfusion reduction with intravenous iron in gynecologic cancer patients receiving chemotherapy. Gynecol. Oncol. 2010, 116, 522–525.
  29. Anthony, L.B.; Gabrail, N.Y.; Ghazal, H.; Woytowitz, D.V.; Flam, M.S.; Drelichman, A.; Loesch, D.M.; Niforos, D.; Mangione, A. IV iron sucrose for cancer and/or chemotherapy-induced anemia in patients treated with erythropoiesis-stimulating agents. Community Oncol. 2011, 8, 270–278.
  30. Auerbach, M.; Ballard, H.; Trout, J.R.; McIlwain, M.; Ackerman, A.; Bahrain, H.; Balan, S.; Barker, L.; Rana, J. 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.
  31. Auerbach, M.; Silberstein, P.T.; Webb, R.T.; Averyanova, S.; Ciuleanu, T.E.; Shao, J.; Bridges, K. Darbepoetin alfa 300 or 500 mug once every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. Am. J. Hematol. 2010, 85, 655–663.
  32. Bastit, L.; Vandebroek, A.; Altintas, S.; Gaede, B.; Pinter, T.; Suto, T.S.; Mossman, T.W.; Smith, K.E.; Vansteenkiste, J.F. 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, 1611–1618.
  33. Hedenus, M.; Birgegard, G.; Nasman, P.; Ahlberg, L.; Karlsson, T.; Lauri, B.; Lundin, J.; Larfars, G.; Osterborg, A. Addition of intravenous iron to epoetin beta increases hemoglobin response and decreases epoetin dose requirement in anemic patients with lymphoproliferative malignancies: A randomized multicenter study. Leukemia 2007, 21, 627–632.
  34. Hedenus, M.; Karlsson, T.; Ludwig, H.; Rzychon, B.; Felder, M.; Roubert, B.; Birgegard, G. Intravenous iron alone resolves anemia in patients with functional iron deficiency and lymphoid malignancies undergoing chemotherapy. Med. Oncol. 2014, 31, 302.
  35. Henry, D.H.; Dahl, N.V.; Auerbach, M.; Tchekmedyian, S.; Laufman, L.R. 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.
  36. Maccio, A.; Madeddu, C.; Gramignano, G.; Mulas, C.; Sanna, E.; Mantovani, G. Efficacy and safety of oral lactoferrin supplementation in combination with rHuEPO-beta for the treatment of anemia in advanced cancer patients undergoing chemotherapy: Open-label, randomized controlled study. Oncologist 2010, 15, 894–902.
  37. Pedrazzoli, P.; Farris, A.; Del Prete, S.; Del Gaizo, F.; Ferrari, D.; Bianchessi, C.; Colucci, G.; Desogus, A.; Gamucci, T.; Pappalardo, A.; et al. Randomized trial of intravenous iron supplementation in patients with chemotherapy-related anemia without iron deficiency treated with darbepoetin alpha. J. Clin. Oncol. 2008, 26, 1619–1625.
  38. Steensma, D.P.; Sloan, J.A.; Dakhil, S.R.; Dalton, R.; Kahanic, S.P.; Prager, D.J.; Stella, P.J.; Rowland, K.M., Jr.; Novotny, P.J.; Loprinzi, C.L. 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, 97–105.
  39. Steensma, D.P.; Sasu, B.J.; Sloan, J.A.; Tomita, D.; Loprinzi, C.L. The relationship between serum hepcidin levels and clinical outcomes in patients with chemotherapy-associated anemia treated in a controlled trial. J. Clin. Oncol. 2011, 29, 9031.
  40. Ansarinejad, N.; Abbasi, B.; Sadat Rasul, M.S.; Fardad, F.; Ramim, T. The Effectiveness of Ferric Carboxymaltose on the Improvement of Chronic Iron Deficiency Anemia in Patients With Colon Cancer: A Controlled Randomized Clinical Trial. J. Obstet. Gynecol. Cancer Res. 2016, 1.
  41. Makharadze, T.; Boccia, R.; Krupa, A.; Blackman, N.; Henry, D.H.; Gilreath, J.A. 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, 1639–1646.
  42. Noronha, V.; Joshi, A.; Patil, V.M.; Banavali, S.D.; Gupta, S.; Parikh, P.M.; Marfatia, S.; Punatar, S.; More, S.; Goud, S.; et al. Phase III randomized trial comparing intravenous to oral iron in patients with cancer-related iron deficiency anemia not on erythropoiesis stimulating agents. Asia Pac. J. Clin. Oncol. 2018, 14, e129–e137.
  43. Dreyer, G.; Mostert, A.E.; Visser, C.; Mouton, A. The severity and optimal management of iron deficiency and anaemia before radiation for cervical cancer at the University of Pretoria Academic Hospitals. South. Afr. J. Gynaecol. Oncol. 2017, 9, 11–15.
  44. Kim, Y.T.; Kim, S.W.; Yoon, B.S.; Cho, H.J.; Nahm, E.J.; Kim, S.H.; Kim, J.H.; Kim, J.W. Effect of intravenously administered iron sucrose on the prevention of anemia in the cervical cancer patients treated with concurrent chemoradiotherapy. Gynecol. Oncol. 2007, 105, 199–204.
  45. Dickson, E.A.; Ng, O.; Keeler, B.D.; Wilcock, A.; Brookes, M.J.; Acheson, A.G. The ICaRAS randomised controlled trial: Intravenous iron to treat anaemia in people with advanced cancer-feasibility of recruitment, intervention and delivery. Palliat Med. 2023, 37, 372–383.
More
Video Production Service