298
Views
17
CrossRef citations to date
0
Altmetric
Review

Iron overload and allogeneic hematopoietic stem-cell transplantation

, &
Pages 71-80 | Published online: 10 Jan 2014

References

  • Majhail NS, Lazarus HM, Burns LJ. Iron overload in hematopoietic cell transplantation. Bone Marrow Transplant.41(12), 997–1003 (2008).
  • Morado M, Ojeda E, Garcia-Bustos J et al. BMT: serum ferritin as risk factor for veno-occlusive disease of the liver. Prospective cohort study. Hematology4(6), 505–512 (2000).
  • Maertens J, Demuynck H, Verbeken EK et al. Mucormycosis in allogeneic bone marrow transplant recipients: report of five cases and review of the role of iron overload in the pathogenesis. Bone Marrow Transplant.24(3), 307–312 (1999).
  • Evens AM, Mehta J, Gordon LI. Rust and corrosion in hematopoietic stem-cell transplantation: the problem of iron and oxidative stress. Bone Marrow Transplant.34(7), 561–571 (2004).
  • Krause A, Neitz S, Magert HJ et al. LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity. FEBS Lett.480(2–3), 147–150 (2000).
  • Park CH, Valore EV, Waring AJ, Ganz T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J. Biol. Chem.276(11), 7806–7810 (2001).
  • Armand P, Kim HT, Cutler CS et al. Prognostic impact of elevated pretransplantation serum ferritin in patients undergoing myeloablative stem-cell transplantation. Blood109(10), 4586–4588 (2007).
  • Neufeld EJ. Oral chelators deferasirox and deferiprone for transfusional iron overload in thalassemia major: new data, new questions. Blood107(9), 3436–3441 (2006).
  • Munoz M, Villar I, Garcia-Erce JA. An update on iron physiology. World J. Gastroenterol.15(37), 4617–4626 (2009).
  • Pigeon C, Ilyin G, Courselaud B et al. A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J. Biol. Chem.276(11), 7811–7819 (2001).
  • Nemeth E, Rivera S, Gabayan V et al. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J. Clin. Invest.113(9), 1271–1276 (2004).
  • Nemeth E, Tuttle MS, Powelson J et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science306(5704), 2090–2093 (2004).
  • Kawabata H, Tomosugi N, Kanda J, Tanaka Y, Yoshizaki K, Uchiyama T. Anti-interleukin 6 receptor antibody tocilizumab reduces the level of serum hepcidin in patients with multicentric Castleman’s disease. Haematologica92(6), 857–858 (2007).
  • Lee P, Peng H, Gelbart T, Wang L, Beutler E. Regulation of hepcidin transcription by interleukin-1 and interleukin-6. Proc. Natl Acad. Sci. USA102(6), 1906–1910 (2005).
  • Kanda J, Uchiyama T, Tomosugi N, Higuchi M, Uchiyama T, Kawabata H. Oncostatin M and leukemia inhibitory factor increase hepcidin expression in hempatoma cell lines. Int. J. Hematol.90(5), 545–552 (2009).
  • Kattamis A, Papassotiriou I, Palaiologou D et al. The effects of erythropoetic activity and iron burden on hepcidin expression in patients with thalassemia major. Haematologica91(6), 809–812 (2006).
  • Kanda J, Mizumoto C, Kawabata H et al. Serum hepcidin level and erythropoietic activity after hematopoietic stem-cell transplantation. Haematologica93(10), 1550–1554 (2008).
  • Sharma S, Nemeth E, Chen YH et al. Involvement of hepcidin in the anemia of multiple myeloma. Clin. Cancer Res.14(11), 3262–3267 (2008).
  • Ganz T, Olbina G, Girelli D, Nemeth E, Westerman M. Immunoassay for human serum hepcidin. Blood112(10), 4292–4297 (2008).
  • Hohaus S, Massini G, Giachelia M et al. Anemia in Hodgkin’s lymphoma: the role of interleukin-6 and hepcidin. J. Clin. Oncol.28(15), 2538–2543 (2010).
  • Shander A, Sazama K. Clinical consequences of iron overload from chronic red blood cell transfusions, its diagnosis, and its management by chelation therapy. Transfusion50(5), 1144–1155 (2010).
  • Tanno T, Bhanu NV, Oneal PA et al. High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Nat. Med.13(9), 1096–1101 (2007).
  • Murphy PT, Mitra S, Gleeson M, Desmond R, Swinkels DW. Urinary hepcidin excretion in patients with low grade myelodysplastic syndrome. Br. J. Haematol.144(3), 451–452 (2009).
  • Winder A, Lefkowitz R, Ghoti H et al. Urinary hepcidin excretion in patients with myelodysplastic syndrome and myelofibrosis. Br. J. Haematol.142(4), 669–671 (2008).
  • Ramirez JM, Schaad O, Durual S et al. Growth differentiation factor 15 production is necessary for normal erythroid differentiation and is increased in refractory anaemia with ring-sideroblasts. Br. J. Haematol.144(2), 251–262 (2009).
  • von Bonsdorff L, Lindeberg E, Sahlstedt L, Lehto J, Parkkinen J. Bleomycin-detectable iron assay for non-transferrin-bound iron in hematologic malignancies. Clin. Chem.48(2), 307–314 (2002).
  • Pietrangelo A. Hereditary hemochromatosis: pathogenesis, diagnosis, and treatment. Gastroenterology139(2), 393–408 (2010).
  • Sahlstedt L, Ebeling F, von Bonsdorff L, Parkkinen J, Ruutu T. Non-transferrin-bound iron during allogeneic stem-cell transplantation. Br. J. Haematol.113(3), 836–838 (2001).
  • Sahlstedt L, von Bonsdorff L, Ebeling F, Parkkinen J, Juvonen E, Ruutu T. Non-transferrin-bound iron in haematological patients during chemotherapy and conditioning for autologous stem-cell transplantation. Eur. J. Haematol.83(5), 455–459 (2009).
  • Weinberg ED. Iron availability and infection. Biochim. Biophys. Acta1790(7), 600–605 (2009).
  • von Bonsdorff L, Sahlstedt L, Ebeling F, Ruutu T, Parkkinen J. Erratum to ‘Apotransferrin administration prevents growth of Staphylococcus epidermidis in serum of stem cell transplant patients by binding of free iron’. [FEMS Immunol. Med. Microbiol.3745–51 (2003)]. FEMS Immunol. Med. Microbiol40(2), 173–180 (2004).
  • Recalcati S, Invernizzi P, Arosio P, Cairo G. New functions for an iron storage protein: the role of ferritin in immunity and autoimmunity. J. Autoimmun.30(1–2), 84–89 (2008).
  • Matzner Y, Hershko C, Polliack A, Konijn AM, Izak G. Suppressive effect of ferritin on in vitro lymphocyte function. Br. J. Haematol.42(3), 345–353 (1979).
  • Broxmeyer HE, Cooper S, Levi S, Arosio P. Mutated recombinant human heavy-chain ferritins and myelosuppression in vitro and in vivo: a link between ferritin ferroxidase activity and biological function. Proc. Natl Acad. Sci. USA88(3), 770–774 (1991).
  • Cardier J, Romano E, Soyano A. Effect of hepatic isoferritins from iron overloaded rats on lymphocyte proliferative response: role of ferritin iron content. Immunopharmacol. Immunotoxicol.17(4), 719–732 (1995).
  • Gray CP, Arosio P, Hersey P. Heavy chain ferritin activates regulatory T cells by induction of changes in dendritic cells. Blood99(9), 3326–3334 (2002).
  • Lee MH, Means RT Jr. Extremely elevated serum ferritin levels in a university hospital: associated diseases and clinical significance. Am. J. Med.98(6), 566–571 (1995).
  • Olive A, Junca J. Elevated serum ferritin levels: associated diseases and clinical significance. Am. J. Med.101(1), 120; author reply 122 (1996).
  • Jastaniah W, Harmatz P, Pakbaz Z, Fischer R, Vichinsky E, Walters MC. Transfusional iron burden and liver toxicity after bone marrow transplantation for acute myelogenous leukemia and hemoglobinopathies. Pediatr. Blood Cancer50(2), 319–324 (2008).
  • St Pierre TG, Clark PR, Chua-Anusorn W et al. Noninvasive measurement and imaging of liver iron concentrations using proton magnetic resonance. Blood105(2), 855–861 (2005).
  • Di Tucci AA, Matta G, Deplano S et al. Myocardial iron overload assessment by T2* magnetic resonance imaging in adult transfusion dependent patients with acquired anemias. Haematologica93(9), 1385–1388 (2008).
  • Emond MJ, Bronner MP, Carlson TH, Lin M, Labbe RF, Kowdley KV. Quantitative study of the variability of hepatic iron concentrations. Clin. Chem.45(3), 340–346 (1999).
  • Sheth S. SQUID biosusceptometry in the measurement of hepatic iron. Pediatr. Radiol.33(6), 373–377 (2003).
  • Anderson LJ, Westwood MA, Holden S et al. Myocardial iron clearance during reversal of siderotic cardiomyopathy with intravenous desferrioxamine: a prospective study using T2* cardiovascular magnetic resonance. Br. J. Haematol.127(3), 348–355 (2004).
  • Tanner MA, He T, Westwood MA, Firmin DN, Pennell DJ. Multi-center validation of the transferability of the magnetic resonance T2* technique for the quantification of tissue iron. Haematologica91(10), 1388–1391 (2006).
  • Angelucci E, Brittenham GM, McLaren CE et al. Hepatic iron concentration and total body iron stores in thalassemia major. N. Engl. J. Med.343(5), 327–331 (2000).
  • Olivieri NF, Brittenham GM. Iron-chelating therapy and the treatment of thalassemia. Blood89(3), 739–761 (1997).
  • Kanda J, Mizumoto C, Ichinohe T et al. Pretransplant serum ferritin and C-reactive protein as predictive factors for early bacterial infection after allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. DOI: 10.1038/bmt.2010.108 (2010) (Epub ahead of print).
  • Armand P, Kim HT, Rhodes J et al. Iron overload in patients with acute leukemia or MDS undergoing myeloablative stem-cell transplantation. Biol. Blood Marrow Transplant. DOI: 10.1016/j.bbmt.2010.09.006 (2010) (Epub ahead of print).
  • Khan FA, Fisher MA, Khakoo RA. Association of hemochromatosis with infectious diseases: expanding spectrum. Int. J. Infect. Dis.11(6), 482–487 (2007).
  • Pullarkat V, Blanchard S, Tegtmeier B et al. Iron overload adversely affects outcome of allogeneic hematopoietic cell transplantation. Bone Marrow Transplant.42(12), 799–805 (2008).
  • Altes A, Remacha AF, Sarda P et al. Early clinical impact of iron overload in stem-cell transplantation. A prospective study. Ann. Hematol.86(6), 443–447 (2007).
  • Spellberg B, Edwards J Jr, Ibrahim A. Novel perspectives on mucormycosis: pathophysiology, presentation, and management. Clin. Microbiol. Rev.18(3), 556–569 (2005).
  • Boelaert JR, Fenves AZ, Coburn JW. Deferoxamine therapy and mucormycosis in dialysis patients: report of an international registry. Am. J. Kidney Dis.18(6), 660–667 (1991).
  • Altes A, Remacha AF, Sarda P et al. Frequent severe liver iron overload after stem-cell transplantation and its possible association with invasive aspergillosis. Bone Marrow Transplant.34(6), 505–509 (2004).
  • Kontoyiannis DP, Chamilos G, Lewis RE et al. Increased bone marrow iron stores is an independent risk factor for invasive aspergillosis in patients with high-risk hematologic malignancies and recipients of allogeneic hematopoietic stem-cell transplantation. Cancer110(6), 1303–1306 (2007).
  • Ozyilmaz E, Aydogdu M, Sucak G et al. Risk factors for fungal pulmonary infections in hematopoietic stem-cell transplantation recipients: the role of iron overload. Bone Marrow Transplant.45(10), 1528–1533 (2010).
  • Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet373(9674), 1550–1561 (2009).
  • Mahindra A, Bolwell B, Sobecks R et al. Elevated pretransplant ferritin is associated with a lower incidence of chronic graft-versus-host disease and inferior survival after myeloablative allogeneic haematopoietic stem-cell transplantation. Br. J. Haematol.146(3), 310–316 (2009).
  • Wahlin A, Lorenz F, Fredriksson M, Remberger M, Wahlin BE, Hagglund H. Hyperferritinemia is associated with low incidence of graft versus host disease, high relapse rate, and impaired survival in patients with blood disorders receiving allogeneic hematopoietic stem cell grafts. Med. Oncol. DOI: 10.1007/s12032-010-9496-1 (2010) (Epub ahead of print).
  • Kataoka K, Nannya Y, Hangaishi A et al. Influence of pretransplantation serum ferritin on nonrelapse mortality after myeloablative and nonmyeloablative allogeneic hematopoietic stem-cell transplantation. Biol. Blood Marrow Transplant.15(2), 195–204 (2009).
  • Busca A, Falda M, Manzini P et al. Iron overload in patients receiving allogeneic hematopoietic stem-cell transplantation: quantification of iron burden by a superconducting quantum interference device (SQUID) and therapeutic effectiveness of phlebotomy. Biol. Blood Marrow Transplant.16(1), 115–122 (2010).
  • Kamble RT, Selby GB, Mims M, Kharfan-Dabaja MA, Ozer H, George JN. Iron overload manifesting as apparent exacerbation of hepatic graft-versus-host disease after allogeneic hematopoietic stem-cell transplantation. Biol. Blood Marrow Transplant.12(5), 506–510 (2006).
  • Lee SH, Yoo KH, Sung KW et al. Hepatic veno-occlusive disease in children after hematopoietic stem-cell transplantation: incidence, risk factors, and outcome. Bone Marrow Transplant.45(8), 1287–1293 (2010).
  • Majhail NS, Lazarus HM, Burns LJ. A prospective study of iron overload management in allogeneic hematopoietic cell transplantation survivors. Biol. Blood Marrow Transplant.16(6), 832–837 (2010).
  • Rose C, Ernst O, Hecquet B et al. Quantification by magnetic resonance imaging and liver consequences of post-transfusional iron overload alone in long term survivors after allogeneic hematopoietic stem-cell transplantation (HSCT). Haematologica92(6), 850–853 (2007).
  • Bair S, Spaulding E, Parkkinen J et al. Transplantation of allogeneic T cells alters iron homeostasis in NOD/SCID mice. Blood113(8), 1841–1844 (2009).
  • Adams PC, Barton JC. How I treat hemochromatosis. Blood116(3), 317–325 (2010).
  • Angelucci E, Muretto P, Lucarelli G et al. Phlebotomy to reduce iron overload in patients cured of thalassemia by bone marrow transplantation. Italian Cooperative Group for Phlebotomy Treatment of Transplanted Thalassemia Patients. Blood90(3), 994–998 (1997).
  • Gaziev D, Giardini C, Angelucci E et al. Intravenous chelation therapy during transplantation for thalassemia. Haematologica80(4), 300–304 (1995).
  • Giardini C, Galimberti M, Lucarelli G et al. Desferrioxamine therapy accelerates clearance of iron deposits after bone marrow transplantation for thalassaemia. Br. J. Haematol.89(4), 868–873 (1995).
  • Nurtjahja-Tjendraputra E, Fu D, Phang JM, Richardson DR. Iron chelation regulates cyclin D1 expression via the proteasome: a link to iron deficiency-mediated growth suppression. Blood109(9), 4045–4054 (2007).
  • Fu D, Richardson DR. Iron chelation and regulation of the cell cycle: 2 mechanisms of posttranscriptional regulation of the universal cyclin-dependent kinase inhibitor p21CIP1/WAF1 by iron depletion. Blood110(2), 752–761 (2007).
  • Donfrancesco A, Deb G, Dominici C, Pileggi D, Castello MA, Helson L. Effects of a single course of deferoxamine in neuroblastoma patients. Cancer Res.50(16), 4929–4930 (1990).
  • Donfrancesco A, De Bernardi B, Carli M et al. Deferoxamine followed by cyclophosphamide, etoposide, carboplatin, thiotepa, induction regimen in advanced neuroblastoma: preliminary results. Italian Neuroblastoma Cooperative Group. Eur. J. Cancer31A(4), 612–615 (1995).
  • Kaloyannidis P, Yannaki E, Sakellari I et al. The impact of desferrioxamine postallogeneic hematopoietic cell transplantation in relapse incidence and disease-free survival: a retrospective analysis. Transplantation89(4), 472–479 (2010).
  • Olivieri NF, Brittenham GM, McLaren CE et al. Long-term safety and effectiveness of iron-chelation therapy with deferiprone for thalassemia major. N. Engl. J. Med.339(7), 417–423 (1998).
  • Pennell DJ, Berdoukas V, Karagiorga M et al. Randomized controlled trial of deferiprone or deferoxamine in β-thalassemia major patients with asymptomatic myocardial siderosis. Blood107(9), 3738–3744 (2006).
  • Cohen AR, Galanello R, Piga A, De Sanctis V, Tricta F. Safety and effectiveness of long-term therapy with the oral iron chelator deferiprone. Blood102(5), 1583–1587 (2003).
  • Cappellini MD, Cohen A, Piga A et al. A phase 3 study of deferasirox (ICL670), a once-daily oral iron chelator, in patients with β-thalassemia. Blood107(9), 3455–3462 (2006).
  • Vichinsky E, Onyekwere O, Porter J et al. A randomised comparison of deferasirox versus deferoxamine for the treatment of transfusional iron overload in sickle cell disease. Br. J. Haematol.136(3), 501–508 (2007).
  • Porter J, Galanello R, Saglio G et al. Relative response of patients with myelodysplastic syndromes and other transfusion-dependent anaemias to deferasirox (ICL670): a 1-yr prospective study. Eur. J. Haematol.80(2), 168–176 (2008).
  • Lee JW, Kang HJ, Kim EK, Kim H, Shin HY, Ahn HS. Effect of iron overload and iron-chelating therapy on allogeneic hematopoietic SCT in children. Bone Marrow Transplant.44(12), 793–797 (2009).
  • Ibrahim AS, Gebermariam T, Fu Y et al. The iron chelator deferasirox protects mice from mucormycosis through iron starvation. J. Clin. Invest.117(9), 2649–2657 (2007).
  • Reed C, Ibrahim A, Edwards JE Jr, Walot I, Spellberg B. Deferasirox, an iron-chelating agent, as salvage therapy for rhinocerebral mucormycosis. Antimicrob. Agents Chemother.50(11), 3968–3969 (2006).
  • Spellberg B, Andes D, Perez M et al. Safety and outcomes of open-label deferasirox iron chelation therapy for mucormycosis. Antimicrob. Agents Chemother.53(7), 3122–3125 (2009).
  • Delea TE, Sofrygin O, Thomas SK, Baladi JF, Phatak PD, Coates TD. Cost effectiveness of once-daily oral chelation therapy with deferasirox versus infusional deferoxamine in transfusion-dependent thalassaemia patients: US healthcare system perspective. Pharmacoeconomics25(4), 329–342 (2007).
  • Tolley K, Oliver N, Miranda E, Migliaccio-Walle K, Bozkaya D, Li Q. Cost effectiveness of deferasirox compared to desferrioxamine in the treatment of iron overload in lower-risk, transfusion-dependent myelodysplastic syndrome patients. J. Med. Econ.13(3), 559–570 (2010).

Website

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.