Skip to main content
Top
Published in: International Urology and Nephrology 2/2012

01-04-2012 | Nephrology – Review

Iron overdose: a contributor to adverse outcomes in randomized trials of anemia correction in CKD

Authors: Peter Van Buren, Ruben L. Velez, Nosratola D. Vaziri, Xin J. Zhou

Published in: International Urology and Nephrology | Issue 2/2012

Login to get access

Abstract

Administration of intravenous iron to supplement erythropoiesis stimulating agents (ESAs) has become a common practice in the management of anemia in patients with end-stage renal disease. Randomized clinical trials of anemia correction in this population have shown more adverse outcomes in CKD and ESRD patients assigned to the higher hemoglobin targets. Retrospective analysis of these trials suggests that morbidity is higher in subjects who fail to achieve the designated hemoglobin target and are typically exposed to higher doses of ESAs and iron than those that easily achieve the intended targets. Intravenous iron administration circumvents the natural biologic mechanisms for handling and utilization of iron. There is in vitro and in vivo evidence that intravenous iron preparations can cause oxidative stress, endothelial dysfunction, inflammation, impaired immunity, and renal injury. Since iron overload is known to promote endothelial dysfunction, cardiovascular disease, and immune dysfunction which are the leading causes of premature mortality in CKD and ESRD patients, it is imperative to exercise caution with the use of IV iron preparations in this population. The present review is intended to provide a brief overview of the potential adverse effects of the overzealous use of these agents.
Literature
1.
go back to reference Vaziri N (2008) Anemia and anemia correction: surrogate markers or causes of mortality in chronic kidney disease. Nat Clin Pract Nephrol 8:436–445CrossRef Vaziri N (2008) Anemia and anemia correction: surrogate markers or causes of mortality in chronic kidney disease. Nat Clin Pract Nephrol 8:436–445CrossRef
2.
go back to reference Vaziri N, Zhou X (2009) Potential mechanisms of adverse outcomes in trials of anemia correction with erythropoietin in chronic kidney disease. Nephrol Dial Transplant 24:1082–1088PubMedCrossRef Vaziri N, Zhou X (2009) Potential mechanisms of adverse outcomes in trials of anemia correction with erythropoietin in chronic kidney disease. Nephrol Dial Transplant 24:1082–1088PubMedCrossRef
3.
go back to reference Ma J, Ebben J, Xia H, Collins A (1999) Hematocrit level and associated mortality in hemodialysis patients. J Am Soc Nephrol 10:610–619PubMed Ma J, Ebben J, Xia H, Collins A (1999) Hematocrit level and associated mortality in hemodialysis patients. J Am Soc Nephrol 10:610–619PubMed
4.
go back to reference Collins A, Li S, Peter W et al (2001) Death, hospitalization, and economic associations among incident hemodialysis patients with hematocrit values of 36–39%. J Am Soc Nephrol 12:2465–2473PubMed Collins A, Li S, Peter W et al (2001) Death, hospitalization, and economic associations among incident hemodialysis patients with hematocrit values of 36–39%. J Am Soc Nephrol 12:2465–2473PubMed
5.
go back to reference Robinson B, Joffe M, Berns J, Pisoni R, Port F, Feldman H (2005) Anemia and mortality in hemodialysis patients: accounting for morbidity and treatment variables updated over time. Kidney Int 68:2323–2330PubMedCrossRef Robinson B, Joffe M, Berns J, Pisoni R, Port F, Feldman H (2005) Anemia and mortality in hemodialysis patients: accounting for morbidity and treatment variables updated over time. Kidney Int 68:2323–2330PubMedCrossRef
6.
go back to reference Locatelli F, Pisoni R, Combe C et al (2004) Anemia in haemodialysis patients of five European countries: association with morbidity and mortality in the dialysis outcomes and practice patterns study (DOPPS). Nephrol Dial Transplant 19:121–132PubMedCrossRef Locatelli F, Pisoni R, Combe C et al (2004) Anemia in haemodialysis patients of five European countries: association with morbidity and mortality in the dialysis outcomes and practice patterns study (DOPPS). Nephrol Dial Transplant 19:121–132PubMedCrossRef
7.
go back to reference Xia H, Ebben J, Ma J, Collins A (1999) Hematocrit levels and hospitalization risks in hemodialysis patients. J Am Soc Nephrol 10:1309–1316PubMed Xia H, Ebben J, Ma J, Collins A (1999) Hematocrit levels and hospitalization risks in hemodialysis patients. J Am Soc Nephrol 10:1309–1316PubMed
8.
go back to reference Li S, Collins A (2004) Association of hematocrit value with cardiovascular morbidity and mortality in incident hemodialysis patients. Kidney Int 65:626–633PubMedCrossRef Li S, Collins A (2004) Association of hematocrit value with cardiovascular morbidity and mortality in incident hemodialysis patients. Kidney Int 65:626–633PubMedCrossRef
9.
go back to reference Ofsthun N, Labrecque J, Lacson E, Keen M, Lazarus J (2003) The effects of higher hemoglobin levels on mortality and hospitalization in hemodialysis patients. Kidney Int 63:1908–1914PubMedCrossRef Ofsthun N, Labrecque J, Lacson E, Keen M, Lazarus J (2003) The effects of higher hemoglobin levels on mortality and hospitalization in hemodialysis patients. Kidney Int 63:1908–1914PubMedCrossRef
10.
go back to reference Wolfe R, Hulbert-Shearon T, Ashby V, Mahadevan S, Port F (2002) Improvements in dialysis patient mortality are associated with improvements in urea reduction ration and hematocrit, 1999–2002. Am J Kidney Dis 45:127–135CrossRef Wolfe R, Hulbert-Shearon T, Ashby V, Mahadevan S, Port F (2002) Improvements in dialysis patient mortality are associated with improvements in urea reduction ration and hematocrit, 1999–2002. Am J Kidney Dis 45:127–135CrossRef
11.
go back to reference NKF-K/DOQI (2006) Clinical practice guidelines for anemia of chronic kidney disease. Am J Kidney Dis 47(Suppl 4):S1 NKF-K/DOQI (2006) Clinical practice guidelines for anemia of chronic kidney disease. Am J Kidney Dis 47(Suppl 4):S1
12.
go back to reference Besarab A, Bolton W, Browne J et al (1998) The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. New Engl J Med 339:584–590PubMedCrossRef Besarab A, Bolton W, Browne J et al (1998) The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. New Engl J Med 339:584–590PubMedCrossRef
13.
go back to reference Drueke T, Locatelli F, Clyne N et al (2006) Normalization of hemoglobin level in patients with chronic kidney disease and anemia. New Engl J Med 355:2071–2084PubMedCrossRef Drueke T, Locatelli F, Clyne N et al (2006) Normalization of hemoglobin level in patients with chronic kidney disease and anemia. New Engl J Med 355:2071–2084PubMedCrossRef
14.
go back to reference Singh A, Szczech L, Tang K et al (2006) Correction of anemia with epoetin alfa in chronic kidney disease. New Engl J Med 355:2085–2098PubMedCrossRef Singh A, Szczech L, Tang K et al (2006) Correction of anemia with epoetin alfa in chronic kidney disease. New Engl J Med 355:2085–2098PubMedCrossRef
15.
go back to reference NKF-K/DOQI (2007) Clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease: 2007 update of hemoglobin target. Am J Kidney Dis 50:474 NKF-K/DOQI (2007) Clinical practice guidelines and clinical practice recommendations for anemia in chronic kidney disease: 2007 update of hemoglobin target. Am J Kidney Dis 50:474
16.
go back to reference Spittle M, Hoenich N, Handelman G, Adhikarla R, Homel P, Levin N (2001) Oxidative stress and inflammation in hemodialysis patients. Am J Kidney Dis 38:1408–1413PubMedCrossRef Spittle M, Hoenich N, Handelman G, Adhikarla R, Homel P, Levin N (2001) Oxidative stress and inflammation in hemodialysis patients. Am J Kidney Dis 38:1408–1413PubMedCrossRef
17.
go back to reference Brownlee M (2001) Biochemistry and molecular cell biology of diabetic complications. Nature 414:813–820PubMedCrossRef Brownlee M (2001) Biochemistry and molecular cell biology of diabetic complications. Nature 414:813–820PubMedCrossRef
18.
go back to reference Schulz E, Gori T, Munzel T (2011) Oxidative stress and endothelial dysfunction in hypertension. Hypertens Res 34:665–673PubMedCrossRef Schulz E, Gori T, Munzel T (2011) Oxidative stress and endothelial dysfunction in hypertension. Hypertens Res 34:665–673PubMedCrossRef
19.
go back to reference Szczech L, Barnhart H, Inrig J et al (2008) Secondary analysis of the CHOIR trial epoetin-alpha dose and achieved hemoglobin outcomes. Kidney Int 74:791–798PubMedCrossRef Szczech L, Barnhart H, Inrig J et al (2008) Secondary analysis of the CHOIR trial epoetin-alpha dose and achieved hemoglobin outcomes. Kidney Int 74:791–798PubMedCrossRef
20.
go back to reference Kuo C, Lee C, Chuang C, Su Y, Chen J (2005) Recombinant human erythropoietin independence in chronic hemodialysis patients: clinical features, iron homeostasis and erythropoiesis. Clin Nephrol 63:92–97PubMed Kuo C, Lee C, Chuang C, Su Y, Chen J (2005) Recombinant human erythropoietin independence in chronic hemodialysis patients: clinical features, iron homeostasis and erythropoiesis. Clin Nephrol 63:92–97PubMed
21.
go back to reference Goodkin DF, Fuller DS, Robinson B et al (2011) Naturally occurring higher hemoglobin concentration does not increase mortality among hemodialysis patients. J Am Soc Nephrol 22:358–365PubMedCrossRef Goodkin DF, Fuller DS, Robinson B et al (2011) Naturally occurring higher hemoglobin concentration does not increase mortality among hemodialysis patients. J Am Soc Nephrol 22:358–365PubMedCrossRef
22.
go back to reference Parfrey P, Foley R, Wittreich B et al (2005) Double-blind comparison of full and partial anemia correction in incident hemodialysis patients without symptomatic heart disease. J Am Soc Nephrol 16:2180–2189PubMedCrossRef Parfrey P, Foley R, Wittreich B et al (2005) Double-blind comparison of full and partial anemia correction in incident hemodialysis patients without symptomatic heart disease. J Am Soc Nephrol 16:2180–2189PubMedCrossRef
23.
24.
go back to reference Jacobs A (1977) Low molecular weight intracellular iron transport compounds. Blood 50:433–439PubMed Jacobs A (1977) Low molecular weight intracellular iron transport compounds. Blood 50:433–439PubMed
25.
go back to reference Burkitt M, Mason R (1991) Direct evidence for in vivo hydroxyl-radical generation in experimental iron overload: An ESR spin-trapping investigation. Proc Natl Acad Sci USA 88:8440–8444PubMedCrossRef Burkitt M, Mason R (1991) Direct evidence for in vivo hydroxyl-radical generation in experimental iron overload: An ESR spin-trapping investigation. Proc Natl Acad Sci USA 88:8440–8444PubMedCrossRef
26.
go back to reference Broedbaek K, Poulsen H, Weimann A et al (2009) Urinary excretion of biomarkers of oxidatively damaged DNA and RNA in hereditary hemochromatosis. Free Radic Biol Med 47:1230–1233PubMedCrossRef Broedbaek K, Poulsen H, Weimann A et al (2009) Urinary excretion of biomarkers of oxidatively damaged DNA and RNA in hereditary hemochromatosis. Free Radic Biol Med 47:1230–1233PubMedCrossRef
27.
go back to reference Kell D (2009) Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics 2:2PubMedCrossRef Kell D (2009) Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics 2:2PubMedCrossRef
28.
go back to reference Beshara S, Lundquist H, Sundin J et al (1999) Kinetic analysis of 52 Fe-labelled iron (III) hydroxide-sucrose complex following bolus administration using positron emission tomography. Br J Hematol 104:288–295CrossRef Beshara S, Lundquist H, Sundin J et al (1999) Kinetic analysis of 52 Fe-labelled iron (III) hydroxide-sucrose complex following bolus administration using positron emission tomography. Br J Hematol 104:288–295CrossRef
29.
go back to reference Zanen A, Adriaansen H, van Bommel E, Posthuma R, de Jong G (1996) Oversaturation of transferrin after intravenous ferric gluconate (ferrlecit) in haemodialysis patients. Nephrol Dial Transplant 11:820–824PubMed Zanen A, Adriaansen H, van Bommel E, Posthuma R, de Jong G (1996) Oversaturation of transferrin after intravenous ferric gluconate (ferrlecit) in haemodialysis patients. Nephrol Dial Transplant 11:820–824PubMed
30.
go back to reference Kooistra M, Kersting S, Lu G et al (2002) Nontransferrin-bound iron in the plasma of the haemodialysis patients after intravenous iron saccharate infusion. Eur J Clin Investig 32(Suppl 1):36–41CrossRef Kooistra M, Kersting S, Lu G et al (2002) Nontransferrin-bound iron in the plasma of the haemodialysis patients after intravenous iron saccharate infusion. Eur J Clin Investig 32(Suppl 1):36–41CrossRef
31.
go back to reference Henderson P, Hillman R (1969) Characteristics of iron dextran utilization in man. Blood 34:357–375PubMed Henderson P, Hillman R (1969) Characteristics of iron dextran utilization in man. Blood 34:357–375PubMed
32.
go back to reference Van Wyck D, Anderson J, Johnson K (2004) Labile iron in parenteral iron formulations: a quantitative and comparative study. Nephrol Dial Transplant 19:561–565PubMedCrossRef Van Wyck D, Anderson J, Johnson K (2004) Labile iron in parenteral iron formulations: a quantitative and comparative study. Nephrol Dial Transplant 19:561–565PubMedCrossRef
33.
go back to reference Scheiber-Mojdehkar B, Sturm B, Plank L, Kryzer I, Goldenberg H (2003) Influence of parenteral iron preparations on non-transferrin bound iron uptake, the iron regulatory protein and the expression of ferritin and the divalent metal transporter DMT-1 in HepG2 human hepatoma cells. Biochem Pharmacol 65:1973–1978PubMedCrossRef Scheiber-Mojdehkar B, Sturm B, Plank L, Kryzer I, Goldenberg H (2003) Influence of parenteral iron preparations on non-transferrin bound iron uptake, the iron regulatory protein and the expression of ferritin and the divalent metal transporter DMT-1 in HepG2 human hepatoma cells. Biochem Pharmacol 65:1973–1978PubMedCrossRef
34.
go back to reference Provenzano R, Schiller B, Rao M et al (2009) Ferumoxytol as an intravenous iron replacement therapy in hemodialysis patients. Clin J Am Soc Nephrol 4:386–393PubMedCrossRef Provenzano R, Schiller B, Rao M et al (2009) Ferumoxytol as an intravenous iron replacement therapy in hemodialysis patients. Clin J Am Soc Nephrol 4:386–393PubMedCrossRef
35.
go back to reference Spinowitz B, Kausz A, Baptista J et al (2008) Ferumoxytol for treating iron deficiency anemia in CKD. J Am Soc Nephrol 19:1599–1605PubMedCrossRef Spinowitz B, Kausz A, Baptista J et al (2008) Ferumoxytol for treating iron deficiency anemia in CKD. J Am Soc Nephrol 19:1599–1605PubMedCrossRef
36.
go back to reference Johnson A, Becker K, Zager R (2010) Parenteral iron formulations differentially affect MCP-1, HO-1, and NGAL gene expression and renal responses to injury. Am J Renal Phys 299:F426–F435CrossRef Johnson A, Becker K, Zager R (2010) Parenteral iron formulations differentially affect MCP-1, HO-1, and NGAL gene expression and renal responses to injury. Am J Renal Phys 299:F426–F435CrossRef
37.
go back to reference Lu M, Suh K, Lee H, Cohen M, Rieves D, Pazdur R (2010) FDA review of ferumoxytol (feraheme) for the treatment of iron deficiency anemia in adults with chronic kidney disease. Am J Hematol 85:315–319PubMed Lu M, Suh K, Lee H, Cohen M, Rieves D, Pazdur R (2010) FDA review of ferumoxytol (feraheme) for the treatment of iron deficiency anemia in adults with chronic kidney disease. Am J Hematol 85:315–319PubMed
38.
go back to reference US Renal Data System (2010) USRDS 2010 annual data report: atlas of chronic kidney disease and end-stage renal disease in the United States. National Institutes of Health, Bethesda US Renal Data System (2010) USRDS 2010 annual data report: atlas of chronic kidney disease and end-stage renal disease in the United States. National Institutes of Health, Bethesda
39.
go back to reference Zager R, Johnson C, Hanson S, Wasse H (2002) Parenteral iron formulations: a comparative toxicologic analysis and mechanisms of cell injury. Am J Kidney Dis 40:90–103PubMedCrossRef Zager R, Johnson C, Hanson S, Wasse H (2002) Parenteral iron formulations: a comparative toxicologic analysis and mechanisms of cell injury. Am J Kidney Dis 40:90–103PubMedCrossRef
40.
go back to reference Rooyakers T, Stroes E, Kooistra M et al (2002) Ferric saccharate induces oxygen radical stress and endothelial dysfunction in vivo. Eur J Clin Investig 32(Suppl 1):9–16CrossRef Rooyakers T, Stroes E, Kooistra M et al (2002) Ferric saccharate induces oxygen radical stress and endothelial dysfunction in vivo. Eur J Clin Investig 32(Suppl 1):9–16CrossRef
41.
go back to reference Roob J, Khoschsorur G, Tiran A et al (2000) Vitamin E attenuates oxidative stress induced by intravenous iron in patients on hemodialysis. J Am Soc Nephrol 11:539–549PubMed Roob J, Khoschsorur G, Tiran A et al (2000) Vitamin E attenuates oxidative stress induced by intravenous iron in patients on hemodialysis. J Am Soc Nephrol 11:539–549PubMed
42.
go back to reference Garcia-Fernandez N, Echevarria A, Sanchez-Ibarrola A, Paramo J, Coma-Cannela I (2010) Randomized clinical trial on acute effects of i.v. iron sucrose during hemodialysis. Nephrology 15:178–183PubMedCrossRef Garcia-Fernandez N, Echevarria A, Sanchez-Ibarrola A, Paramo J, Coma-Cannela I (2010) Randomized clinical trial on acute effects of i.v. iron sucrose during hemodialysis. Nephrology 15:178–183PubMedCrossRef
43.
go back to reference Salahudeen A, Oliver B, Bower J, Roberts L (2001) Increase in plasma esterified F2-isoprostanes following intravenous iron infusion in patients on hemodialysis. Kidney Int 60:1525–1531PubMedCrossRef Salahudeen A, Oliver B, Bower J, Roberts L (2001) Increase in plasma esterified F2-isoprostanes following intravenous iron infusion in patients on hemodialysis. Kidney Int 60:1525–1531PubMedCrossRef
44.
go back to reference Kuo K, Hung S, Wei Y, Tarng D (2008) Intravenous iron exacerbates oxidative DNA damage in peripheral blood lymphocytes in chronic hemodialysis patients. J Am Soc Nephrol 19:1817–1826PubMedCrossRef Kuo K, Hung S, Wei Y, Tarng D (2008) Intravenous iron exacerbates oxidative DNA damage in peripheral blood lymphocytes in chronic hemodialysis patients. J Am Soc Nephrol 19:1817–1826PubMedCrossRef
45.
go back to reference Tovbin D, Mazour D, Voroblov M, Chalmovitz C, Meyerstein N (2002) Induction of protein oxidation by intravenous iron in hemodialysis patients: role of inflammation. Am J Kidney Dis 40:1005–1012PubMedCrossRef Tovbin D, Mazour D, Voroblov M, Chalmovitz C, Meyerstein N (2002) Induction of protein oxidation by intravenous iron in hemodialysis patients: role of inflammation. Am J Kidney Dis 40:1005–1012PubMedCrossRef
46.
go back to reference Agarwal R, Vasavada N, Sachs N, Chase S (2004) Oxidative stress and renal injury with intravenous iron in patients with chronic kidney disease. Kidney Int 65:2279–2289PubMedCrossRef Agarwal R, Vasavada N, Sachs N, Chase S (2004) Oxidative stress and renal injury with intravenous iron in patients with chronic kidney disease. Kidney Int 65:2279–2289PubMedCrossRef
47.
go back to reference Kielstein J, Boger R, Bode-Boger S (1999) Asymmetric dimethyl arginine concentrations differ in patients with end stage renal disease: relationship to treatment method and atherosclerotic disease. J Am Soc Nephrol 10:594–600PubMed Kielstein J, Boger R, Bode-Boger S (1999) Asymmetric dimethyl arginine concentrations differ in patients with end stage renal disease: relationship to treatment method and atherosclerotic disease. J Am Soc Nephrol 10:594–600PubMed
48.
go back to reference Zoccali C, Bode-Boger S, Mallamaci F et al (2001) Plasma concentrations of asymmetrical dimethyl arginine and mortality in patients with end-stage renal disease: a prospective study. Lancet 358:2113–2117PubMedCrossRef Zoccali C, Bode-Boger S, Mallamaci F et al (2001) Plasma concentrations of asymmetrical dimethyl arginine and mortality in patients with end-stage renal disease: a prospective study. Lancet 358:2113–2117PubMedCrossRef
49.
go back to reference Matsuguma K, Ueda S, Yamagishi S et al (2006) Molecular mechanisms for elevation of asymmetric dimethyl arginine and its role for hypertension in chronic kidney disease. J Am Soc Nephrol 17:2176–2183PubMedCrossRef Matsuguma K, Ueda S, Yamagishi S et al (2006) Molecular mechanisms for elevation of asymmetric dimethyl arginine and its role for hypertension in chronic kidney disease. J Am Soc Nephrol 17:2176–2183PubMedCrossRef
50.
go back to reference Kartikasari A, Georgiou N, Visseren F, van Kats-Renaud H, van Sweder A, Marx J (2006) Endothelial activation and induction of monocyte adhesion by nontransferrin-bound iron present in human sera. FASEB J 20:353–355PubMed Kartikasari A, Georgiou N, Visseren F, van Kats-Renaud H, van Sweder A, Marx J (2006) Endothelial activation and induction of monocyte adhesion by nontransferrin-bound iron present in human sera. FASEB J 20:353–355PubMed
51.
go back to reference Schaller G, Scheibert-Mohdehkar B, Wolzt M et al (2005) Intravenous iron increases labile serum iron but does not impair blood flow reactivity in dialysis patients. Kidney Int 68:2814–2822PubMedCrossRef Schaller G, Scheibert-Mohdehkar B, Wolzt M et al (2005) Intravenous iron increases labile serum iron but does not impair blood flow reactivity in dialysis patients. Kidney Int 68:2814–2822PubMedCrossRef
52.
go back to reference Reis K, Guz G, Ozdemir H et al (2005) Intravenous iron therapy as a possible risk factor for atherosclerosis in end stage renal disease. Int Heart J 46:255–264PubMedCrossRef Reis K, Guz G, Ozdemir H et al (2005) Intravenous iron therapy as a possible risk factor for atherosclerosis in end stage renal disease. Int Heart J 46:255–264PubMedCrossRef
53.
go back to reference Duffy S, Biegelsen E, Holbrook M et al (2001) Iron chelation improves endothelial cell function in patients with coronary artery disease. Circulation 103:2799–2804PubMed Duffy S, Biegelsen E, Holbrook M et al (2001) Iron chelation improves endothelial cell function in patients with coronary artery disease. Circulation 103:2799–2804PubMed
54.
go back to reference Zager R (2005) Parenteral iron treatment induces MCP-1 accumulation in plasma, normal kidneys, and in experimental nephropathy. Kidney Int 68:1533–1542PubMedCrossRef Zager R (2005) Parenteral iron treatment induces MCP-1 accumulation in plasma, normal kidneys, and in experimental nephropathy. Kidney Int 68:1533–1542PubMedCrossRef
55.
go back to reference Agarwal R (2006) Proinflammatory effects of iron sucrose in chronic kidney disease. Kidney Int 69:1259–1263PubMedCrossRef Agarwal R (2006) Proinflammatory effects of iron sucrose in chronic kidney disease. Kidney Int 69:1259–1263PubMedCrossRef
56.
go back to reference Weiss G, Meusberger E, Radacher G, Garimorth K, Neyer U, Myaer G (2003) Effect of iron treatment on circulating cytokine levels in ESRD patients receiving recombinant human erythropoietin. Kidney Int 64:572–578PubMedCrossRef Weiss G, Meusberger E, Radacher G, Garimorth K, Neyer U, Myaer G (2003) Effect of iron treatment on circulating cytokine levels in ESRD patients receiving recombinant human erythropoietin. Kidney Int 64:572–578PubMedCrossRef
57.
go back to reference Sarnak M, Jaber B (2000) Mortality caused by sepsis in patients with end-stage renal disease compared with the general population. Kidney Int 58:1758–1764PubMedCrossRef Sarnak M, Jaber B (2000) Mortality caused by sepsis in patients with end-stage renal disease compared with the general population. Kidney Int 58:1758–1764PubMedCrossRef
58.
go back to reference Kato S, Chmielewski M, Honda H et al (2008) Aspects of immune dysfunction in end-stage renal disease. Clin J Am Soc Nephrol 3:1526–1533PubMedCrossRef Kato S, Chmielewski M, Honda H et al (2008) Aspects of immune dysfunction in end-stage renal disease. Clin J Am Soc Nephrol 3:1526–1533PubMedCrossRef
59.
go back to reference Djeha A, Brock J (1992) Uptake and intracellular handling of iron from transferrin and iron chelates by mitogen stimulated mouse lymphocytes. Biochem Biopys Acta 1133:147–152CrossRef Djeha A, Brock J (1992) Uptake and intracellular handling of iron from transferrin and iron chelates by mitogen stimulated mouse lymphocytes. Biochem Biopys Acta 1133:147–152CrossRef
60.
go back to reference Deicher R, Ziai F, Cohen G, Mullner M, Horl W (2003) High dose parenteral iron sucrose depresses neutrophil intracellular killing capacity. Kidney Int 64:728–736PubMedCrossRef Deicher R, Ziai F, Cohen G, Mullner M, Horl W (2003) High dose parenteral iron sucrose depresses neutrophil intracellular killing capacity. Kidney Int 64:728–736PubMedCrossRef
61.
go back to reference Guo D, Jaber B, Lee S et al (2002) Impact of iron dextran on polymorphonuclear cell function among hemodialysis patients. Clin Nephrol 58:134–142PubMed Guo D, Jaber B, Lee S et al (2002) Impact of iron dextran on polymorphonuclear cell function among hemodialysis patients. Clin Nephrol 58:134–142PubMed
62.
go back to reference Gupta A, Zhuo J, Zha J, Reddy S, Olp J, Pai A (2010) Effect of different intravenous iron preparations on lymphocyte intracellular reactive oxygen species generation and subpopulation survival. BMC Nephrol 17:11–16 Gupta A, Zhuo J, Zha J, Reddy S, Olp J, Pai A (2010) Effect of different intravenous iron preparations on lymphocyte intracellular reactive oxygen species generation and subpopulation survival. BMC Nephrol 17:11–16
63.
go back to reference Tenopoulou M, Doulias P, Barbouti A, Brunk U, Galaris D (2005) Role of compartmentalized redox-active iron in hydrogen peroxide-induced DNA damage and apoptosis. Biochem J 387:703–710PubMedCrossRef Tenopoulou M, Doulias P, Barbouti A, Brunk U, Galaris D (2005) Role of compartmentalized redox-active iron in hydrogen peroxide-induced DNA damage and apoptosis. Biochem J 387:703–710PubMedCrossRef
64.
go back to reference Zager R, Johnson A, Hanson S (2004) Parenteral iron nephrotoxicity: potential mechanisms and consequences. Kidney Int 66:144–156PubMedCrossRef Zager R, Johnson A, Hanson S (2004) Parenteral iron nephrotoxicity: potential mechanisms and consequences. Kidney Int 66:144–156PubMedCrossRef
65.
go back to reference Agarwal R (2005) On the nature of proteinuria with acute renal injury in patients with chronic kidney disease. Am J Physiol Renal Physiol 288:F265–F271PubMedCrossRef Agarwal R (2005) On the nature of proteinuria with acute renal injury in patients with chronic kidney disease. Am J Physiol Renal Physiol 288:F265–F271PubMedCrossRef
66.
go back to reference Agarwal R, Rizkala A, Kaskas M, Minasian R, Trout J (2007) Iron sucrose causes greater proteinuria than ferric gluconate in non-dialysis chronic kidney disease. Kidney Int 72:638–642PubMedCrossRef Agarwal R, Rizkala A, Kaskas M, Minasian R, Trout J (2007) Iron sucrose causes greater proteinuria than ferric gluconate in non-dialysis chronic kidney disease. Kidney Int 72:638–642PubMedCrossRef
67.
go back to reference Leehey D, Palubiak D, Chebrolu S, Agarwal R (2005) Sodium ferric gluconate causes oxidative stress but not acute renal injury in patients with chronic kidney disease: a pilot study. Nephrol Dial Transplant 20:135–140PubMedCrossRef Leehey D, Palubiak D, Chebrolu S, Agarwal R (2005) Sodium ferric gluconate causes oxidative stress but not acute renal injury in patients with chronic kidney disease: a pilot study. Nephrol Dial Transplant 20:135–140PubMedCrossRef
Metadata
Title
Iron overdose: a contributor to adverse outcomes in randomized trials of anemia correction in CKD
Authors
Peter Van Buren
Ruben L. Velez
Nosratola D. Vaziri
Xin J. Zhou
Publication date
01-04-2012
Publisher
Springer Netherlands
Published in
International Urology and Nephrology / Issue 2/2012
Print ISSN: 0301-1623
Electronic ISSN: 1573-2584
DOI
https://doi.org/10.1007/s11255-011-0028-5

Other articles of this Issue 2/2012

International Urology and Nephrology 2/2012 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.