Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

REVIEWS

Iron supplementation to treat anemia


in patients with chronic kidney disease
Anatole Besarab and Daniel W. Coyne
Abstract | Iron deficiency is prevalent in patients with chronic kidney disease (CKD), and use of oral and
intravenous iron in patients with CKD who do not require dialysis might obviate or delay the need for treatment
with eythropoiesis-stimulating agents (ESAs). Patients on hemodialysis have lower intestinal iron absorption,
greater iron losses, and require greater iron turnover to maintain the ESA-driven red cell mass than do healthy
individuals. In these patients, intravenous iron reduces ESA dose requirements and increases the likelihood
of maintaining levels of hemoglobin within the desired range. Oral iron is inferior to intravenous iron in
patients on hemodialysis, in part because elevated serum levels of hepcidin prevent intestinal absorption
of iron. Increased levels of hepcidin also impair the normal recycling of iron through the reticuloendothelial
system. Levels of serum ferritin and transferrin saturation below 450 pmol/l and 20%, respectively are
indicative of iron deficiency, but values above the normal range lack diagnostic value in patients with CKD on
dialysis. The availability of various iron preparations and new developments in delivering iron should enable
adequate provision of iron to patients with CKD. This Review examines the efficacy, safety and use of iron
supplementation therapy for the treatment of anemia in patients with CKD.
Besarab, A. & Coyne, D. W. Nat. Rev. Nephrol. 6, 699–710 (2010); published online 19 October 2010; corrected online 12 December 2011;
doi:10.1038/nrneph.2010.139

Introduction
Compared with healthy individuals, individuals with differentiation and survival of these cells.5,6 The duration
end-stage renal disease (ESRD) demonstrate major of this erythropoietin-dependent first stage varies from
differ­ences in iron metabolism: decreased duodenal iron 8 days to 13 days and shortens as erythrocyte production
absorption, decreased iron transport capacity because of increases. Each erythroblast ultimately produces about
a reduced transferrin concentration, increased iron loss 32 daughter cells that leave the bone marrow as reticulo­
owing to frequent blood sampling, occult gastrointestinal cytes complete with their full content of hemoglobin. This
bleeding and other means (such as losses of blood into second stage of rapid cell division and hemoglobinization
dialysis tubing and dialyzers), and an increased rate of takes about 4 days. Virtually all of the iron needed to gen-
iron turnover to maintain the decreased red blood cell erate a hemoglobin-laden reticulocyte is taken up within
mass (Figure 1).1 At hemoglobin levels of 100–120 g/l, 2–3 days during this stage (Figure 2). Clearly, therefore,
red blood cell losses associated with hemodialysis equate the treatment of anemia in patients with CKD must both
to iron losses of 6–7 mg daily in addition to physio­ promote the production of erythroblasts and ensure that
logical iron losses of 1–2 mg daily. With the additional iron levels are adequate to enable optimal hemoglobin
losses from periodic laboratory evaluations, annual formation in the daughter cells.
iron losses can exceed 1.5–3.0 g.2 Occult blood loss con- Anemia was thought to increase the incidence of
tributes to anemia in patients with advanced CKD who cardio­vascular events and the risk of death in patients
are not yet on hemodialysis, and as many as 25–45% of with CKD. Since the development of erythropoiesis-
such patients might be iron-deficient.3,4 stimulating agents (ESAs) in the late 1980s (discussed in Division of Nephrology
and Hypertension,
During erythropoiesis, committed erythroid progeni- detail elsewhere7), anemia in patients with CKD has been Henry Ford Hospital,
tors (burst-forming unit-erythroid [BFU‑E] and colony- treated with these agents to reduce the need for blood CFP 511, 2799 West
forming unit-erythroid [CFU‑E]) appear sequentially and transfusions, improve health-related quality of life and Grand Boulevard,
Detroit, MI 48301, USA
mature morphologically into recognizable erythroblasts to decrease the risk of cardiovascular events and death. (A. Besarab). Division
(Figure 2). Erythropoietin is critical for multiplica­tion, The dose of ESAs needed to achieve adequate levels of Nephrology,
Washington University
of hemo­globin varies widely from <1,000 U to 30,000 U School of Medicine,
Competing interests per hemo­dialysis treatment. The dose and frequency of 660 South Euclid
A. Besarab declares associations with the following companies: ESA administration is, therefore, usually flexible. Findings Avenue, St Louis,
MO 63110, USA
Affymax, AMAG Pharmaceuticals, Amgen, Hoffman–La Roche, from randomized trials indicate that ESAs improve (D. W. Coyne).
Watson Pharmaceuticals. D. W. Coyne declares associations
quality of life and cardiovascular outcomes when used
with the following companies: AMAG Pharmaceuticals, Correspondence to:
Pharmacosmos, Sanofi–Aventis, Watson Pharmaceuticals. See to treat patients with hemoglobin levels that correspond to A. Besarab
the article online for full details of the relationships. moderate anemia (90–120 g/l). However, ESA therapy in abesara1@hfhs.org

NATURE REVIEWS | NEPHROLOGY VOLUME 6  |  DECEMBER 2010  |  699


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points that use of intravenous iron can lower ESA dose require-
■■ Iron deficiency is prevalent in patients with chronic kidney disease (CKD) and
ments in patients with CKD who are on dialysis.10,11 As the
should be treated increased cardio­vascular risk associated with ESA therapy
■■ Inconsistent and inadequate absorption of oral iron frequently leads to the
might be a result of the high doses of these agents rather
need for administration of intravenous iron than of the elevation in hemo­globin levels, iron supplemen-
■■ Hepcidin (levels of which are frequently increased in patients with CKD) impairs
tation might reduce the risks associ­ated with ESA therapy.
iron absorption and the normal recycling of iron through the reticuloendothelial Nevertheless, the long-term safety of iron administration
system has not been thoroughly tested in patients with CKD. This
■■ Low levels of serum ferritin and transferrin saturation indicate iron deficiency, Review examines the efficacy, safety and rational use of
but values in or above the normal range lack diagnostic value iron therapy for the treatment of anemia in patients with
■■ Frequent, small repeat doses of intravenous iron are more effective than CKD. Although ESA therapy is mentioned throughout this
infrequent, large doses for maintaining levels of hemoglobin and reducing the Review, these agents are not the main focus of this article
requirement for erythropoiesis-stimulating agents (ESAs) and as such will not be discussed in detail as they have been
■■ Relative thrombocytosis (owing to iron deficiency) might contribute to the comprehensively reviewed elsewhere.7,12
increased cardiovascular risk seen with high ESA doses
Iron deficiency anemia and CKD
Iron dynamics
Reticuloendothelial stores
ferritin level: 33–450 pmol/l
Iron absorption from food seems to be unimpaired in
ferritin level: 110–1,800 pmol/l patients with CKD in whom levels of ferritin (a protein
32 mg per day that stores iron) are not elevated.13,14 Eschbach and co-
workers13,14 observed that the proportion of iron absorbed
Transferrin from food in the gastrointestinal tract of patients with
Transferrin saturation uremia varied inversely with the logarithm of serum
Tissue 200 mg Gut
15–50% (men), levels of ferritin. The researchers concluded that uremia
12–45% (women)
TSAT 20% did not interfere with physiological iron absorption but
36 mg per day Loss: 1 mg per day their findings clearly indicated that the amount of iron
absorbed from dietary sources might not be suffi­cient
Red blood cells to meet the requirements for erythropoiesis in these
2,050–2,500 mg Net loss through
(hematocrit 0.41–0.50) hemodialysis: patients. Findings from a 1984 study (before ESA therapy
1,750 mg
(hematocrit 0.35)
4.5–6.0 mg per day became available) indicated normal iron uptake by red
blood cells as a result of oral administration of ferrous
Figure 1 | Iron distribution in the average adult. Values for healthy individuals are
sulfate with ascorbic acid in fasting patients.15 However,
shown in black print whereas those for patients with end-stage renal disease
(ESRD) on hemodialysis are shown in red print. The overall balance of iron in
the extent of absorption varied widely (4–30% of a 50 mg
patients on chronic maintenance hemodialysis is impaired. Transport of iron is oral dose of ferrous sulfate, mean 13%). Only one-third of
decreased by a reduction in circulating transferrin levels. At the same time, dialysis- the patients had normal iron incorporation into red blood
associated blood losses lead to loss of iron from the body. Daily incorporation of cells after this treatment, and none showed an improve-
iron into red blood cell hemoglobin and its release from senescent cells is actually ment in their hematocrit. By contrast, intravenous iron
increased in patients with ESRD on hemodialysis compared with healthy individuals. therapy increased both iron uptake by red blood cells
Note that both occult blood loss in stool and loss of red blood cells in tubing and hematocrit.14 Although these results indicate that
increase the daily loss of iron in patients with ESRD on hemodialysis.90
iron utilization is decreased in patients on dialysis who
are not receiving ESAs, even with ESA use iron utiliza-
patients with hemoglobin levels >120 g/l reduces the need tion remains suboptimal, particularly in the presence of
for blood transfusions, but increases the risk of adverse inflammation. In some patients, low levels of transferrin,
cardiovascular events.8,9 A full discussion of the ongoing which result in a low capacity for binding iron, impair the
controversy with regard to the level of hemoglobin that metabolic cycling of iron in patients without CKD.
should prompt ESA therapy is beyond the scope of this Acquired gastrointestinal disease is increasingly
Review. However, we do believe that avoidance of blood recog­nized as an additional malabsorptive mechanism
transfusion in renal transplant candi­dates is important of iron deficiency anemia in patients on hemodialysis:
to prevent sensitization owing to the formation of pre- auto­immune atrophic gastritis and Helicobacter pylori
formed reactive antibodies, and recommend the initiation infection are frequently present in such patients. 16,17
of ESA therapy in these patients when the level of hemo- Endoscopic abnormalities consisting of chronic and
globin is <100 g/l rather than <90 g/l. Iron therapy can be acute gastritis, duodenitis or duodenal ulcer gastritis are
initiated in any patient with symptoms of iron deficiency found in up to 90% of asymptomatic patients on hemo-
irrespective of their level of hemoglobin. dialysis.18 Moreover, histological examination of multiple
Moderate anemia is frequently observed in patients with antral gastric biopsy samples documented the presence
nondialysis-dependent CKD (NDD-CKD) who are not of chronic active gastritis in these patients.18
receiving ESA therapy. Our clinical experience has shown
that use of oral and intravenous iron in such patients can Inflammation
increase their hemoglobin levels—delaying or obviating Patients with CKD often exhibit chronic inflammation.
the need for ESAs. Results from studies also demonstrate Furthermore, uremia is considered an inflammatory

700  |  DECEMBER 2010  |  VOLUME 6  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

CD34
Stem Progenitor cells Precursor cells
cells (BFU-E, CFU-E) (erythroblasts) Reticulocytes

GM-CSF
IGF-I Apoptosis
IL-3
SCF

Neocytolysis
(spleen)

Erythropoietin-dependent

~8–13 days 4 days

Figure 2 | Erythropoiesis. Erythropoietin is required in the first stage of erythropoiesis (in which multipotent stem cells form
progenitor BFU-E and CFU-E cells) but not in the second stage (in which precursor cells form erythroblasts and reticulocytes).
The interval from stem cell to erythroblast is ~17 days, with 8–13 days spent in the BFU and CFU stages. Erythropoietin acts
on BFU-E and CFU-E cells for approximately 7–10 days. Sites of action of erythropoietin and other growth factors during the
stages of erythropoiesis are shown. The stippling indicates potential apoptosis of progenitor cells. The erythropoietin-
independent phase of erythropoiesis begins with the erythroblast and ends with the released reticulocyte. Each erythroblast
produces a progeny of 32 cells, which must synthesize the appropriate amount of hemoglobin before they are released into
the circulation. Reticulocytes released into the circulation undergo volume surface area remodeling but are subject to
neocytolysis (premature death, which only occurs when erythropoietin levels decline abruptly below a critical level) for up
to 10 days as they traverse the spleen. Abbreviations: BFU-E, burst-forming unit-erythroid; CFU-E, colony-forming unit-
erythroid; GM-CSF, granulocyte–macrophage colony-stimulating factor; IGF-I, insulin-like growth factor I; IL-3, interleukin 3;
SCF, stem cell factor. Permission obtained from Wolters Kluwer © Besarab, A. & Yee, J. in Principles and Practice of Dialysis,
4th edn Ch. 30 (ed. Henrich, W. L.) 499–523 (Lippincott Williams & Wilkins, Baltimore, 2009).

state even in the absence of apparent infection or obvious The role of hepcidin
inflammatory conditions. Early observational clinical In many patients with CKD, poor absorption of dietary
studies associated high serum ferritin levels with infec- iron and an inability to use the body’s iron stores con-
tion19,20 or mortality.21 Hyperferritinemia-associated tribute to anemia. These abnormalities in iron balance
morbidity could be related to inflammatory processes result from increased levels of the hormone hepcidin (a
that are not associated with iron, and high serum ferritin 25 amino acid peptide synthesized in hepatocytes that is
levels might merely be a marker of acute-phase infection a key regulator of iron levels). Hepcidin is released into
or inflammation,22 which would make the association the circulation and binds to solute family 40 member 1,
between hyperferritinemia and increased risk of infec- which is also termed ferroportin 1. After binding to
tion and death a mere epiphenomenon. Indeed, levels of target cells (chiefly enterocytes and tissue macrophages),
the inflammatory cytokine tumor necrosis factor were the hepcidin–ferroportin 1 complex is internalized and
decreased in patients on dialysis who received intra­ degraded, which leads to decreased iron efflux from
venous iron compared to those in patients who did not iron-exporting tissues into plasma. By this mechanism,
receive this therapy. 23 In the same vein, Feldman and hepcidin inhibits dietary iron absorption, the efflux of
co-workers (who initially noted a tendency towards an recycled iron from splenic and hepatic macrophages, and
increased risk of death in patients on dialysis who were the release of iron from storage in hepatocytes.26
receiving high doses of intravenous iron)24 revised their Hepcidin synthesis is stimulated by excessive plasma
conclusions when they used models that adjusted for bias iron and iron stores and inhibited by increased erythro-
by indication.25 poietic activity. The results of a 2010 study indicated that

NATURE REVIEWS | NEPHROLOGY VOLUME 6  |  DECEMBER 2010  |  701


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

a b Enucleating erythoblast c

Reticulocyte Macrophage Reticulocyte

Figure 3 | Photomicrographs of erythroblastic islands, where erythroid precursors proliferate, differentiate and enucleate.
The interaction of cells within the erythroblastic island is essential for both early and late stages of erythroid maturation.
a | Conventional photomicrograph of an erythroblastic island. b | Scanning electron microscopy image of an erythroblastic
island. Macrophages phagocytose extruded erythroblast nuclei at the end stage of erythroid maturation.
c | Photomicrograph showing immunofluorescence staining of an erythroblastic island. Central macrophages can be
identified by their unique immunophenotypic signature. The macrophages are adhesive and function as nurse cells. Both
macrophages and erythroblasts display adhesive interactions that maintain island integrity. Such interactions enable
regulatory feedback within islands and also trigger intracellular signaling pathways that regulate gene expression for
cellular growth within the erythroblastic islands. Permission obtained from the American Society of Hematology ©
Chasis, J. A. & Mohandas, M. Blood 112, 470–478 (2008).

erythroblasts produce a circulating factor that inhibits functional. The rate of iron delivery to the bone marrow
production of hepcidin and maximizes the delivery of is determined by the amount of iron in the circulation,
iron to the erythron.27 As bone marrow erythropoiesis where it is bound to the iron-transporting protein trans-
occurs in erythroblastic islands that surround macro­ ferrin. As transferrin concentration is reduced by 30%
phages (Figure 3),28 hepcidin inhibition facilitates the in patients with ESRD, functional iron deficiency can
mobilization of iron stored in the reticuloendothelial develop in such patients because the demands of the
system in macrophages to the surrounding maturing ESA-stimulated bone marrow for iron outstrip the supply
red blood cells. capacity of transferrin.
The role of abnormalities in hepcidin synthesis in Previous studies of iron status in patients with renal
anemia associated with renal disease and in resistance diseases discriminated between absolute and functional
to erythropoietic therapies remains unclear because iron deficiency.32,33 Absolute iron deficiency reflects
of the difficulty in measuring levels of the biologically minimal to no stores of iron, and is thought to be present
active form of this hormone and the direct influence of when transferrin saturation and ferritin levels are both
inflamma­tion on hepcidin synthesis.29 Currently available low (for example, <20% and <449 pmol/ml, respectively).
immunoassays mostly measure levels of pro­hepcidin. Functional iron deficiency describes the inadequate
Until 2 years ago, immunoassays to measure serum release of iron to support the needs of erythropoiesis,
levels of hepcidin did not measure the intact hormone. despite the presence of adequate stores of iron, and
However, even when hepcidin level is measured using a typically occurs when erythropoiesis is driven by ESA
reliable immunoassay of the intact hormone30 or by mass therapy in patients with inflammation. Functional iron
spectroscopy, variation in hepcidin levels within the same deficiency is thought to be present when serum levels of
individual might reflect not only changes in iron state ferritin are high but transferrin saturation is low. Studies
and changes in renal elimination owing to kidney failure over the past 5 years, however, undermine this dichoto-
but also changes that occur because of inflammation. mous view. Although patients on dialysis without ade-
Inflammation increases hepcidin concentrations, quate iron stores are likely to also have low serum levels of
leading to iron sequestration in macrophages, which ferritin, some of these individuals have elevated levels
results in low levels of circulating iron and eventually of serum ferritin, and a few patients also have trans­ferrin
anemia. The fact that serum hepcidin levels are closely saturation values >20%. Additionally, many patients
linked to systemic inflammation might limit the useful- with CKD who have adequate levels of stored iron have
ness of this marker in assessing iron status in patients with iron-restricted erythropoiesis, which can be successfully
CKD. Whether diagnosis of different forms of anemia will treated with intravenous iron therapy.34,35
be facilitated by improved hepcidin assays or whether Discriminating between absolute and functional iron
anemia treatment will be enhanced by the develop­ment deficiency is difficult in clinical practice without histo­
of hepcidin agonists and antagonists remains unclear.31 logical examination of the bone marrow (Figure 4).
For these reasons, hepcidin will not be further discussed Although patients with absolute iron deficiency are more
in this Review as an indicator of iron status. likely than those with functional deficiency to respond
to intravenous iron, about 30% of patients with func-
Absolute and functional iron deficiency tional iron deficiency will respond to such treatment.
Iron deficiency is a frequent finding in patients who Consequently, in patients on hemodialysis who have
have anemia associated with renal disease, particularly iron-restricted erythropoiesis, a trial of intravenous iron
in those receiving ESAs. As these agents enable the pro- therapy is recommended when hemoglobin is below target
duction of supraphysiologic levels of red blood cells, iron levels or high ESA doses are used, and serum ferri­tin
deficiency in these patients can be either absolute or and transferrin saturation values are equivocal.

702  |  DECEMBER 2010  |  VOLUME 6  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

In a trial of patients with NDD-CKD, Stancu and


colleagues35 found that the proportion of patients who
responded to intravenous iron was higher among patients
with iron-deficient bone marrow than among those with
iron-replete bone marrow (63% versus 30%). Peripheral iron
indices, such as a transferrin saturation of 15% and a serum
ferritin level of 169 pmol/l had a moderate accuracy (15%
transferrin saturation, positive predictive value 76% and
negative predictive value 72%; 169 pmol/l serum ferritin
level, positive predictive value 74% and negative predictive
value 70%) in predicting a 10 g/l increase in hemoglobin
in response to intra­venous iron therapy. The chances of a
positive response to intravenous iron therapy increased
by 7% for each 1% decrease in transferrin saturation.36 As
an erythropoietic response was observed in one-third of Figure 4 | Microphotograph of bone marrow staining for
patients with an iron-replete bone marrow, and periph- iron. Iron (blue) is located mainly in the macrophages
eral iron indices had only a moderate utility in predict- (arrow, lower left). Image courtesy of P. Beris, Athens
ing a response to iron supplementation, the researchers University, Greece.
concluded that a test to predict a patient’s response to
intravenous iron therapy would be a useful tool in the The choice of iron therapy depends both on the stage
management of anemia in patients with NDD-CKD. of renal disease and, if the patient is on dialysis, the
dialysis modality chosen. The use of intravenous iron
Diagnosis was avoided for nearly half a century as it was consid-
Iron deficiency should be diagnosed by a global examina­ ered potentially dangerous, and its use was restricted to
tion of the patient’s symptoms and appropriate laboratory patients who could not tolerate oral iron supplementa-
investigations. Inflammation or infection can elevate tion. This attitude changed with the advent of ESAs to
serum ferritin levels and reduce trans­ferrin levels, and treat CKD-associated anemia, especially in patients on
malnutrition can also reduce transferrin levels. Trans­ hemodialysis. The introduction of various iron formula-
ferrin is also a nutritional marker and levels of this tions, such as the low-molecular-weight iron dextrans
protein are as powerful a predictor of death as serum ferrous gluconate and iron sucrose (also known as iron
albumin levels.37 Consequently, a transferrin saturation saccharate), which are safer than the previously available
value within the normal range could mask functional agent, high-molecular-weight iron dextran, has reduced
iron deficiency and the need for iron therapy. A low ferri- the clinician’s concerns for the risk of anaphylaxis. Except
tin level (for example, <450 pmol/l in patients with CKD among patients treated with high-molecular-weight
on hemodialysis or <225 pmol/l in patients with NDD- iron dextrans, which are still available, serious or life-
CKD) is a reliable indicator of absolute iron deficiency, threatening adverse events of intravenous iron seem to
and such patients generally require treatment. A moder- be rare.
ately high serum ferritin level (>1,123 pmol/l), however, The management of iron deficiency anemia in patients
does not exclude iron deficiency or assure replete iron with NDD-CKD is controversial, particularly with regard
stores.38 Decisions on the use of iron supplementation to the use of oral versus intravenous iron supplementa-
in patients with high serum levels of ferritin should be tion. In practice, oral administration of iron (for example,
driven by the severity of anemia, the use and dose of ESA, as iron sulfate, iron fumarate or heme) is preferred in
and the history of iron administration. patients with NDD-CKD because of its convenience and
to avoid systemic adverse events such as hypotension or
Treatment options for anemia anaphylactoid reactions.
Many patients with CKD and anemia (irrespective of
whether they are on dialysis) do not respond adequately Oral iron therapy
to anemia treatment (that is, reach target levels of hemo- The primary source of iron in the diet is heme and its
globin) or require high doses of ESA and iron. As men- absorption is efficiently regulated such that its accumula­
tioned earlier, use of high ESA doses in patients with tion by the gut is prevented when iron is supplied in
hemoglobin levels >120 g/l is associated with increased excess.39 In patients with CKD, iron absorption seems
cardiovascular morbidity and mortality, particularly to be normal in the absence of any underlying gastric
when high-dose ESAs are used in individuals who or duodenal pathology,13 but ingestion of certain foods
respond poorly to treatment. Iron deficiency is the major and drugs can interfere with oral iron absorption.
impediment to the cost-effective use of ESAs. Conversely, Furthermore, absorption of iron salts, such as ferrous
iron supplementation without ESA treatment is usually sulfate, can be influenced both positively and negatively
unsuccessful, particularly in patients on hemodialysis. by various amino acids.40 Iron salts should, therefore, be
For these reasons, most patients with advanced CKD taken on an empty stomach, although this practice might
require concurrent ESA and iron administration to treat increase gastric symptoms and hinder patients’ compli-
CKD-associated anemia efficiently. ance with therapy. Proton pump inhibitors that alter

NATURE REVIEWS | NEPHROLOGY VOLUME 6  |  DECEMBER 2010  |  703


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

duodenal pH and the conversion of iron from Fe2+ to levels fall to <450 pmol/l, with a subsequent pulse dose of
Fe3+ also interfere with iron absorption. Other commonly 1 g iron, which is administered over the course of several
prescribed medications, such as calcium acetate, lantha- hemodialysis treatments.
num carbonate, and aluminum-containing ant­a cids In a summary of seven studies of presumably iron-
also impair iron absorption. replete patients on hemodialysis, administration of a
In the ESA era, oral iron supplementation should be weekly dose of intravenous iron increased hematocrit by
used whenever possible in patients with NDD-CKD 14% and reduced epoetin alfa dose by 38%.46 In another
although on occasion intravenous iron will be needed study, administration of 6.25–21.3 mg of ferrous glucon-
in those with ongoing slow blood losses caused by ate at every hemodialysis session was more effective in
disease or treatment with anticoagulant or antiplatelet increasing hemoglobin and maintaining stable serum
drugs. By contrast, an early study indicated that oral ferritin levels than 62.5 mg of ferrous gluconate every
iron supplementation was rarely successful in patients 1–4 weeks.47 Different intravenous iron regimens have
on hemo­dialysis.41 In this study, the participants received been investigated in patients on hemodialysis—a ‘main-
~200 mg of elemental iron daily for 6 months using one tenance’ regimen of 25–100 mg of iron every 1–2 weeks
of four iron preparations in conjunction with 100 mg (to maintain transferrin saturation >20% and serum
ascorbic acid. The group that achieved the highest trans­ levels of ferritin >450 pmol/l) versus a traditional ‘load
ferrin saturation values (that is, >20%) during the study and hold’ regimen of intermittent repletion of 1 g of iron
period were those treated with ferrous fumarate. Another (administered only when transferrin saturation was
double-­blind, placebo-controlled study evaluated the <20% or serum ferritin levels were <450 pmol/l). Patients
efficacy of oral iron administration in 49 patients on who received the maintenance regimen required 50%
hemodialysis.42 Iron-replete patients randomly assigned less ESAs overall than did those in the other group to
to oral iron supplementation showed signs of depleted maintain the same levels of hemoglobin.10 Surprisingly,
iron stores similar to that observed in patients assigned the cumulative amount of iron needed over a 72-week
to placebo. Oral (nonheme) iron is usually inadequate for period was identical for the two regimens. When two
maintenance of iron stores in patients on hemodialysis, different threshold values for transferrin were used
but heme-containing polypeptides are of interest as they (20–30% versus 30–50%), a further 40% reduction in the
could overcome some of the limitations of traditional ESA dose was achieved, although 2–3 times more iron
oral iron preparations.43 Ofalabi et al.44 compared the was transiently needed to achieve the higher of the two
efficacy of 200 mg per day iron fumarate versus 24 mg per transferrin values.11
day heme-containing polypeptide and found no differ­ In contrast with the 33 days needed for orally admin-
ences in either the efficacy or the profile of adverse effects istered iron to reach maximal red blood cell incorpora-
between the two treatments.44 In summary, no specific tion alluded to above, the time to maximal red blood cell
oral iron preparation can be definitively recommended incorporation for intravenous iron dextran was 8.6 days
over another. (within the normal time frame of 4–10 days). 15 After
The ineffectiveness of oral iron preparations in patients 3 months of either oral or intravenous iron therapy in
on hemodialysis might result from problems with iron patients on hemodialysis, hemoglobin levels remained
incorporation into red blood cells. A small study (before unchanged in the oral-therapy group whereas those in the
the advent of ESA therapy) in 14 patients on hemo­ intravenous-therapy group increased.15 Intravenous iron
dialysis compared the kinetics of an 59Fe isotope as well therefore seems to be better utilized than orally admin-
as whole-body 59Fe counts following administration of istered iron in patients on maintenance hemo­dialysis
radiolabeled iron, either intravenously as iron dextran or who are not treated with ESAs. The use of ESAs should
orally as iron sulfate.15 Following oral iron supplementa- magnify this difference. Whether regular supple­mentation
tion, iron absorption was normal (13% of dose) but the with oral iron could reduce the amount of intravenous
time required to reach maximal red cell incorporation iron needed in ESA-treated patients is unknown.
was prolonged to 33 days.15
Safety
Intravenous iron therapy Intravenous iron administration was viewed with appre-
Each intravenous iron product available obtained hension in the pre-ESA era and as a consequence was
approval for clinical use because it increased hemoglobin little used in the management of anemia in patients with
levels better than oral iron supplements or controls. The CKD. Before ESAs became available, the risk and conse-
response of hemoglobin to intravenous iron is related to quences of hemochromatosis as a result of blood trans­
the amount of iron administered, with 1 g of iron gener- fusion or iron administration to patients with anemia on
ally considered an adequate test of iron responsiveness. dialysis were frequently reported. To observe the effects
Patients with adequate levels of iron who received peri- of hemochromatosis on parenchymal organs, serum
odic intravenous iron supplementation maintained better ferri­tin levels had to exceed 4,500–6,700 pmol/l, and were
serum ferritin levels and had lower requirements for commonly >11,300 pmol/l.48
ESAs than did patients who received no intra­venous iron The iron storage capacity of the reticulo­endothelial
or just oral iron.45 In traditional inter­mittent (loading) system is ~5 g. The risk of iron overload increases
dose regimens, intravenous iron is given only when when transferrin saturation is persistently >50%, and
transferrin saturation is <20% and/or serum ferri­t in iron overload with parenchymal iron deposition is

704  |  DECEMBER 2010  |  VOLUME 6  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

a 25 – b 10,000 –

9,000 –

20 – 8.000 –

Serum ferritin level (pmol/l)


7,000–
Number of patients

15 – 6,000 –

5,000 –

10 – 4,000 –

3,000 –

5– 2.000 –

1,000–

0– 0–
0 200 2,000 20,000 200,000 Negative 1+ 2+ 3+ 4+
Ferritin (pmol/l) Hepatosplenic iron scores
Figure 5 | Serum ferritin level and reticuloendothelial iron accumulation in patients on hemodialysis treated for anemia with
blood transfusions in the pre-ESA era. a | Serum ferritin levels in 120 patients on maintenance hemodialysis.50 Only 15 of
patients had serum ferritin <100 pmol/l, the remaining patients had levels 2,250–45,000 pmol/l. Average iron overload was
8.8 g accrued over 3.6 years (the storage capacity of the reticuloendothelial system is ~5 g). Postmortem analysis showed
that 16 of 22 patients with serum ferritin levels >2,250 pmol/l had tissue that stained positive for iron; all had iron in the
reticuloendothelial system, one had iron in hepatocytes, and four patients had iron in cardiac myocytes. None had fibrosis.
b | Correlation of serum ferritin concentration with semiquantitative scores for hepatosplenic iron deposits in 36 patients on
hemodialysis.49 Reticuloendothelial staining for iron increases in proportion with serum ferritin concentrations. Hepatic iron
level had been maintained <22 mg/kg of body weight (the threshold that induces hepatic damage) even after 7–8 years of
repeated blood transfusions (data not shown). Risk of damage (fibrosis) from intravenous iron administration was low
(<20%), damage was mild, and only occurred in patients with a 4+ score of bone marrow iron. Median serum ferritin levels in
these patients was 5,620 pmol/l, markedly higher than the current recommended values of 1,120–1,800 pmol/l.
Abbreviation: ESA, erythropoiesis-stimulating agent. Permission for panel a obtained from Oxford University Press ©
Gokal, R. et al. Q. J. Med. 48, 369–391 (1979) and for panel b from Elsevier Ltd © Ali, M. Lancet 1, 652–655 (1982).

character­ized by very high serum ferritin levels (typi- with a second dose 3–7 days later.58,59 Ferumoxytol can
cally >4,494 pmol/l) and transferrin saturation >70%.49 also occasionally cause anaphylactoid reactions. Phase
Before the availability of ESAs, iron accumulation III trials of this drug in patients with CKD excluded
occurred in patients with CKD on dialysis owing to patients with iron allergies or more than two drug aller-
repeated blood transfusions. These patients typically gies, as such patients seem to be at an increased risk of
had serum ferritin levels >4,494 pmol/l (Figure 5), high severe drug reactions.58,59 Ferric carboxy­maltose can be
levels of iron in macrophages in the liver and spleen, but administered as a series of 200 mg injections, or as a 1 g
very little parenchymal iron accumulation (in hepato- infusion.60 This last iron product is approved in Europe,
cytes or cardiac myocytes).49,50 Only three of 16 patients but not in the USA owing to safety concerns (because of
on dialysis with ferritin levels >2,247 pmol/l had iron in a high incidence of anaphylactoid reactions and deaths).
parenchymal cells. By contrast, none had fibrosis and all The FDA has requested additional safety data to be
16 had abundant levels of iron in the reticuloendothelial obtained from clinical studies, which are presently being
system.50 Moderately high levels of serum ferritin (that conducted in the USA. Caution and careful monitoring
is, ≤4,500 pmol/l) were not associated with excessive should, however, be used when administering any intra-
iron storage in tissues in a postmortem examination of venous iron product and the risks of potential adverse
indivi­duals who were on hemodialysis.51,52 reactions should be balanced against the benefits.
However, all intravenous iron products can lead
to acute adverse reactions, which can be minor to life Mechanisms of harm
threatening. The low-molecular-weight iron dextrans The high level of oxidative stress present in most patients
seem to have a markedly lower incidence of associated with CKD on hemodialysis61 has been hypothesized to be
severe anaphylactoid reactions than high-molecular- exacerbated by the administration of iron, and in particu-
weight iron dextrans.53,54 Nevertheless, all such agents are lar intravenous iron. This hypothesis proposes that iron
associated with life-threatening anaphylaxis. Ferric glu- administration at doses well below those associated with
conate55 and iron sucrose56 have a reduced incidence of parenchymal iron overload increases the risk of infec-
anaphylactoid reactions, but can cause hypotension with tions, cardiovascular events and death. Findings from
increasing doses. Consequently, these two iron prod- in vitro studies indicate that iron supplementation could
ucts must be given as several divided doses to provide increase markers of oxidative stress.62 Early observa-
1 g of iron, the standard dose needed to treat anemia.57 tional clinical studies associated iron administra­tion with
Ferumoxytol can be administered as a 510 mg injection, indices of cardiovascular disease63 or risk of death24 in

NATURE REVIEWS | NEPHROLOGY VOLUME 6  |  DECEMBER 2010  |  705


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

patients on hemodialysis. The redox balance could have Heart Association) included 41% of patients with an esti-
a substantial pathogenetic role in these processes through mated glomerular filtration rate <60 ml/min/1.73 m2.73
the regulation of signaling pathways, gene expression, cell In these patients, intravenous iron improved symptoms,
proliferation and fibrosis. Oxidative stress is involved in functional capacity (as measured by a 6 min walking test)
uremia-related inflammation64 and experimental evi- and quality of life. Among the patients randomly allo-
dence indicates that labile iron might change the redox cated to receive intravenous iron therapy, no increase in
state to produce increased oxidative stress.65 However, the incidence of infections, hospitalizations, or deaths
interpretation of the contribution of intravenous iron was observed versus placebo. Trials have yet to find
therapy to these processes is confounded by the observa­ definitive evidence to support the hypothesis that use
tion that hemodialysis itself, even with so-called bio­ of intravenous iron is harmful in patients with CKD on
compatible membranes, produces an imbalance between hemo­dialysis. Two large, randomized, ongoing trials will
the production of reactive oxygen species and anti­oxidant address the important question of whether intravenous
mechanisms.66 The observation by Drüeke et al.63 that or oral iron supplementation affects the progression of
intima–media thickness of the common carotid artery renal dys­function in patients with CKD.74,75
and wall-to-lumen thickness ratio were associated with Administration of ESAs can result in increased platelet
plasma levels of advanced oxida­tion protein products, counts, because these agents can induce iron deficiency,
serum levels of ferritin and the annual intravenous iron which has long been known to increase the platelet
dose administered are hypothesis-generating only, and count. The increased number of thrombovascular and
provide no proof of causality.63 Although of interest, dis- cardiovascular events associated with ESA therapy
cussion of whether iron chelators could provide protection might, in part, result from increased platelet counts.
against the effects of labile iron67 is premature. Co-administration of iron would prevent this increase
Subsequent observational studies found that the and thus reduce the risk of thrombovascular and cardio­
increased mortality in patients on hemodialysis was vascular events. A randomized trial of iron in oncology
largely linked to high serum ferritin levels,21 owing to the patients receiving ESAs found lower platelet counts and
confounding effects of malnutrition and inflammation.68 a lower rate of thrombovascular events in patients treated
A low, rather than a high, serum iron level was strongly with iron supplementation than in patients who did not
associated with high mortality and hospitalization in receive this therapy.76 Streja et al.77 found that a high
patients on hemodialysis. Similarly, high trans­ferrin platelet count was associated with low iron stores and
saturation values were associated with low mortality in with prescription of high doses of recombinant human
patients with NDD-CKD who were under the care of erythropoietin in a large population of patients on
the US administration’s Veterans Affairs department.69 hemodialysis. Data from the DRIVE trial in patients
Lastly, the relationship of iron use to death in patients on hemodialysis indicated that platelet counts decreased
on hemodialysis occurs only at extremely high annual significantly in patients who received intravenous iron
doses of iron (that is, >4,800 mg per year), and can be a (by a mean of 29,000 per μl, P = 0.017), but remained
manifestation of confounding by indication.68 Finally, unchanged in patients who were not given iron. Our
a 2009 observational study of 1,774 patients on main­ research group reported a mean decrease in the platelet
tenance hemodialysis who were followed up for 9.5 years count of 50,000 per μl in patients with iron deficiency
concluded that long-term survival was favorably affected who were not receiving dialysis and were being treated
by indicators of iron sufficiency (serum ferritin level with intravenous iron.78 Alternatively, thrombocytosis in
>1,350 pmol/l and transferrin saturation >25%) and by ESA-treated patients on hemodialysis might be mediated
moderate iron administration.70 by ESA rather than by iron deficiency.79 In patients with
A number of randomized trials have assessed the safety normal kidney function who have iron deficiency, correc­
of iron administration in patients with CKD, but all are tion of the deficiency reduces the platelet count. This
of relatively short duration and small size compared to reduction is associated with changes in levels of endo­
the trials of ESA in this setting.71–75 In short-term studies, genous erythropoietin but not in levels of other platelet
single doses of iron sucrose (but not of ferric gluconate) cytokines, such as thrombopoietin, leukemia inhibitory
induced transient proteinuria in patients with NDD- factor, interleukin 6 and interleukin 11. Obtaining data
CKD, although the clinical relevance of this finding is on platelet counts from participants in large trials of
unknown.71 The DRIVE (Dialysis Patients’ Response iron therapy for anemia would be of great interest and
to Intravenous Iron with Elevated Ferritin) study,72 a would enable the relationships between platelet counts,
random­ized trial in which patients on hemodialysis (with iron deficiency, intravenous iron administration and
serum ferritin levels of 1,124–2,696 pmol/ml and trans- cardiovascular events to be examined in detail.
ferrin saturation values ≤25%) were treated with high
doses of epoetin alfa (>30,000 U per week), found that Factors in the choice of iron therapy
1 g of intravenous iron did not increase the risk of infec- Intravenous iron can produce a marked increase in the
tion and resulted in reduced numbers of serious adverse hemoglobin level (to 120 g/l) of some patients with CKD
events compared with those who had no iron over the and iron-deficiency anemia, without ESA therapy.80 Some
3 months of observation. A 6-month, double-blind patients with NDD-CKD who remain anemic despite
trial of intravenous iron versus placebo in patients with treatment with oral iron supplementation for 5–6 weeks
class II and III heart failure (according to the New York subsequently respond to intravenous iron therapy.58

706  |  DECEMBER 2010  |  VOLUME 6  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Patients with NDD-CKD and anemia Patients with CKD and anemia on dialysis

Patients on hemodialysis Patients on CAPD


Do patients have an iron-replete state?
■ Transferrin saturation >25%
■ Serum ferritin level >225 pmol/l

Yes No

Treatment No treatment Maintenance iron therapy:


with ESAs with ESAs ■ Intravenous iron supplementation is
more costly than oral iron therapy, but
the cost differential might decrease
over time because of the ESA-sparing
effect of intravenous iron therapy
Maintenance iron therapy: ■ Low-dose intravenous iron Maintenance iron therapy:
■ To achieve transferrin saturation >30% (25–62 mg weekly) ■ Relatively low ongoing blood losses
and serum ferritin level >450 pmol/l ■ Transferrin saturation maintained at compared with patients on hemodialysis
■ Oral iron supplementation preferable >25% ■ Intravenous iron administration of
to intravenous iron ■ Stable serum ferritin levels at 500 mg or 1 g of iron as a slow infusion
■ ‘Disguising’ oral iron therapy by 450–1,800 pmol/l over 4–6 h maintains adequate levels
prescribing it in multivitamin formulation ■ Additional intravenous iron administered of hemoglobin for 6 months to 1 year
increases patients’ adherence to patients who require high ESA dose ■ Alternatively, monthly doses of
■ Addition of decussate to oral iron or have a hemoglobin level that is 100–250 mg iron can be administered
formulation decreases constipation persistently below target intravenously at each visit to the clinic

Patients on proton pump inhibitors, antiplatelet agents or anticoagulants usually


develop hyporesponsiveness to ESAs owing to iron deficiency
■ Administer cumulative dose of 1 g iron intravenously
■ Low-molecular-weight dextran as a single 500 mg or 1 g dose infused over 1.5–4 h
■ Patients monitored in clinic
■ Patients with ongoing gastrointestinal bleeding might require up to three 1 g infusions
of iron per year

Figure 6 | A treatment algorithm for the management of anemia in patients with CKD. Clinical decision support systems are
used within the Henry Ford Health Care System and patients with NDD-CKD and anemia are treated according to a
computerized anemia-management program. Abbreviations: CAPD, continuous ambulatory peritoneal dialysis; CKD, chronic
kidney disease; ESA, erythropoiesis-stimulating agent; NDD-CKD, nondialysis-dependent CKD.

A meta-analysis of studies published between 1970 and The amount of iron required to maximize the hemo-
200981 concluded that iron supplementation was advis- globin response and repletion of iron stores can only be
able for all iron-deficient patients with CKD who were estimated by the severity of anemia, taking into account
receiving ESA therapy—consistent with FDA recom- indications of whether iron stores are low. Patients
mendations. Another meta-analysis identified 13 trials should receive the amount of iron needed to correct the
(6 of which included patients with CKD and 7 of which red blood cell mass, usually as 75–125 mg intravenous
included patients on hemodialysis) in which compari- weekly doses, until the desired total amount is given. We
sons of efficacy and safety assessments were made over a calculate the amount of iron required on the basis that
period of up to 2 months.60 In patients on hemodialysis, 1 mg of iron is needed to achieve each 1 ml increase in the
those treated with intravenous iron therapy had consider­ amount of packed red blood cells. The increase in packed
ably greater hemoglobin levels than those treated with red blood cells is estimated from the starting and target
oral iron (weighted mean difference, 8.3 g/l). Increases in hematocrit and the estimated blood volume of the patient
the hemoglobin level were positively associated with the (70–80 ml/kg body weight). Thus, the required amount
dose of intravenous iron. For patients with CKD, a sub- in mg of iron is calculated as (desired hematocrit–­initial
stantial difference (but smaller than that observed with hematocrit) × 75. Using this calcula­t ion, therefore,
patients on hemodialysis) in hemoglobin level favored 750 mg of iron would be needed to obtain an increase
the intravenous iron group (weighted mean difference in the patient’s hematocrit from 24% to 34%. In patients
3.1 g/l). Although data for all-cause mortality were who are iron depleted, additional iron is need to account
sparse, no differences in adverse events were observed for ongoing blood losses occurring during the period of
in the groups of patients who received intravenous iron correction (about 3 months), estimated as 600–700 mg.
versus oral iron. In each of four randomized trials that When the total amount of iron required exceeds 1 g,
included patients with NDD-CKD who were not treated iron replenishment is best achieved by administration
with ESAs, the hemoglobin response was greater with of 100–125 mg of iron three times a week. Patients’ iron
intravenous than with oral iron.60 Iron supplementation status and levels of serum ferritin and transferritin
can avoid or delay the need for ESA therapy in some must be monitored at monthly intervals to enable the
patients with NDD-CKD.82 cost-effective management of anemia with epoetin alfa.

NATURE REVIEWS | NEPHROLOGY VOLUME 6  |  DECEMBER 2010  |  707


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

In the Henry Ford Health Care System, clinical deci- a week. Findings from another study in patients with
sion support systems are used to provide treatment high ESA requirements showed that administration of
recom­mendations for an individual patient. In particular, 300 mg of ascorbic acid with each dialysis treatment
patients with NDD-CKD and anemia are treated accord- over 6 months increased hemoglobin levels and reduced
ing to a computerized anemia-management program.83 C‑reactive protein levels from 190 nmol/l to 95 nmol/l.
Longitudinal data on patients’ transferrin saturation and A trend towards decreased levels of serum ferritin was
serum ferritin levels are entered into this program, also evident in the treated group, whereas hemoglobin
and used to direct their treatment (Figure 6). and iron parameters remained unchanged in the control
group.88 In another study, patients with iron overload
Novel methods of iron delivery but not aluminum overload were treated with desferri­
New preparations that permit rapid, high-dose oxamine mesilate.89 The dose of epoetin alfa required
administra­t ion of iron could change the outpatient decreased dramatically from 400 U/kg to 25 U/kg and
management of iron deficiency anemia. In a random- hemoglobin levels increased to 110 g/l.89
ized study, a single 15 min injection of ferric carboxy­
maltose, repeated up to twice if required, resulted in Conclusions
53.2% of patients with NDD-CKD achieving a ≥10 g/l Neither any specific value for transferrin saturation nor
increase in hemoglobin level by day 56 without ESAs, the upper limit for serum ferritin levels (<2,700 pmol/l)
compared with only 29.9% of patients who were given can assure adequate delivery of iron to the erythron
oral iron supple­ments.84 Administration of two 510 mg during ESA therapy. Suboptimal responses to ESA
ferumoxytol injections about 1 week apart increased the therapy are commonly caused by the failure to supply
mean hemoglobin level by 10 g/l compared with a <2.0 g/l adequate amounts of iron. This functional iron defi-
increase in hemoglobin levels in patients randomly ciency can only be discerned by a response to challenge
assigned to receive oral iron supplementation.85 with intravenous iron therapy. The decision to use intra-
Administration of iron via the dialyzate during each venous iron therapy versus oral iron supplementation
hemodialysis session is also undergoing testing. In a should be made on the basis of the individual patient’s
preliminary 6‑month study of stable patients on hemo­ overall condition. If the benefits outweigh the risks, a
dialysis, soluble ferric pyrophosphate (a chelated iron) trial of intravenous iron therapy might be warranted.
added to the dialyzate readily diffused into the blood The typical patient who merits a trial of intravenous iron
compartment and was safe and effective.86 supplementation is without evidence of active infection
Occasionally, patients clearly have adequate iron and has a history of failure to achieve target hemoglobin
stores but respond poorly even to large ESA doses. In levels despite administration of large doses of ESA.
such circumstances, several options exist. Ascorbic
acid (500 mg) administered intravenously after dialysis
Review criteria
1–3 times weekly can mobilize tissue iron stores.87 The
patients’ serum ferritin levels remained unchanged with We searched PubMed for papers in the English language
this treatment, but their transferrin saturation increased published between 1970 and June 2009. Search terms
from 27% to 54% and their hematocrit increased from included “iron metabolism” and “chronic kidney disease”.
27% to 32%. As ascorbic acid is metabolized to oxalate, Only full-text papers were chosen for discussion in this
which is removed poorly by dialysis, we recommend Review. Our extensive collection of papers was also
searched for further suitable articles.
that no more than 300 mg be administered three times

1. Bothwell, T. et al. Iron Metabolism in Man 2nd edn with cardiac disease. N. Engl. J. Med. 339, 15. Magana, L., Dhar, S. K., Smith, E. C. &
(Blackwell Scientific Publishing, 1979). 584–590 (1998). Martinez, C. Iron absorption and utilization in
2. Sakiewicz, P. & Paganini, E. The use of iron in 9. Fishbane, S. & Besarab, A. Mechanisms of maintenance hemodialysis patients: oral and
patients on chronic dialysis: mistake and increased mortality risk with erythropoietin intravenous routes. Mt Sinai J. Med. 51,
misconceptions. J. Nephrol. 11, 5–15 (1998). treatment to higher hemoglobin targets. Clin. J. 180–183 (1984).
3. Rabiner, S. F. Uremic bleeding. Prog. Hemost. Am. Soc. Nephrol. 2, 1274–1282 (2007) 16. Hershko, C. & Skikne, B. Pathogenesis and
Thromb. 1, 233–250 (1972). 10. Besarab, A., Kaiser, J. W. & Frinak, S. management of iron deficiency anemia:
4. Powell, N. & McNair, A. Gastrointestinal A study of parenteral iron regimens in emerging role of celiac disease, Helicobacter
evaluation of anaemic patients without evidence hemodialysis patients. Am. J. Kidney Dis. 34, pylori, and autoimmune gastritis. Semin.
of iron deficiency. Eur. J. Gastroenterol. Hepatol. 21–28 (1999). Hematol. 46, 339–350 (2009).
20, 1094–1100 (2008). 11. Besarab, A. et al. Optimization of epoetin therapy 17. Sugimoto, M., Sakai, K., Kita, M., Imanishi, J. &
5. Gregory, C. G. & Eaves, A. C. Three stages of with intravenous iron therapy in hemodialysis Yamaoka, Y. Prevalence of Helicobacter pylori
erythropoietic progenitor cell differentiation patients. J. Am. Soc. Nephrol. 11, 530–538 infection in long-term hemodialysis patients.
distinguished by a number of physical and (2000). Kidney Int. 75, 96–103 (2009).
biological properties. Blood 51, 527–537 12. Wish, J. B. Past, present, and future of chronic 18. Al-Mueilo, S. H. Gastroduodenal lesions and
(1978). kidney disease anemia management in the Helicobacter pylori infection in hemodialysis
6. Jelkmann, W. & Hellwig-Burgel, T. Biology of United States. Adv. Chronic Kidney Dis. 16, patients. Saudi Med. J. 25, 1010–1014 (2004).
erythropoietin. Adv. Exp. Med. Biol. 502, 101–108 (2009). 19. Waterlot, Y. et al. Impaired phagocytic activity of
169–187 (2001). 13. Eschbach, J. W., Cook, J. D. & Finch, C. A. Iron neutrophils in patients receiving haemodialysis:
7. Besarab, A. & Yee, J. in Principles and Practice of absorption in chronic renal disease. Clin. Sci. 38, the critical role of iron overload. Br. Med. J. (Clin.
Dialysis (ed. Henrich, W. L.) 499–523 (Lippincott, 191–201 (1970). Res. Ed.) 291, 501–504 (1985).
Williams and Wilkens, Baltimore, 2009). 14. Eschbach, J. W., Cook, J. D., Scribner, B. H. & 20. Seifert, A., von Herrath, D. & Schaefer, K. Iron
8. Besarab, A. et al. The effects of normal versus Finch, C. A. Iron balance in hemodialysis overload, but not treatment with desferrioxamine
anemic hematocrit on hemodialysis patients patients. Ann. Int. Med. 87, 710–713 (1977). favours the development of septicemia in

708  |  DECEMBER 2010  |  VOLUME 6  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

patients on maintenance hemodialysis. Q. J. 40. Tonini, M., D’Angelo, L., Carminati, G. M. & 60. Macdougall, I. C. Evolution of IV iron compounds
Med. 65, 1015–1024 (1987). De Bernardi, M. Intestinal absorption of iron. I. over the last century. J. Ren. Care 35 (Suppl. 2),
21. Kalantar-Zadeh, K., Don, B. R., Rodriguez, R. A. Interference of amino acids and polypeptides 8–13 (2009).
& Humphreys, M. H. Serum ferritin is a marker [Italian]. Boll. Chim. Farm. 118, 49–52 (1979). 61. Himmelfarb, J., Stenvinkel, P., Ikizler, T. A. &
of morbidity and mortality in hemodialysis 41. Wingard, R. L., Parker, R. A., Ismail, N. & Hakim, R. M. The elephant in uremia: oxidant
patients. Am. J. Kidney Dis. 37, 564–572 Hakim, R. M. Efficacy of oral iron therapy in stress as a unifying concept of cardiovascular
(2001). patients receiving recombinant human disease in uremia. Kidney Int. 62, 1524–1538
22. Kalantar-Zadeh, K., Rodriguez, R. A. & erythropoietin. Am. J. Kidney Dis. 25, 433–439 (2002).
Humphreys, M. H. Association between serum (1995). 62. Zager, R. A., Johnson, A. C., Hanson, S. Y. &
ferritin and measures of inflammation, nutrition 42. Markowitz, G. S., Kahn, G. A., Feingold, R. E., Wasse, H. Parenteral iron formulations:
and iron in haemodialysis patients. Nephrol. Coco, M. & Lynn, R. I. An evaluation of the a comparative toxicologic analysis and
Dial. Transplant. 19, 141–149 (2004). effectiveness of oral iron therapy in hemodialysis mechanisms of cell injury. Am. J. Kidney Dis. 40,
23. Weiss, G. et al. Effect of iron treatment on patients receiving recombinant human 90–103 (2002).
circulating cytokine levels in ESRD patients erythropoietin. Clin. Nephrol. 48, 34–40 (1997). 63. Drüeke, T. et al. Iron therapy, advanced oxidation
receiving recombinant human erythropoietin. 43. Nissenson, A. R. et al. Clinical evaluation of protein products, and carotid artery intima-media
Kidney Int. 64, 572–578 (2003). heme iron polypeptide: sustaining a response to thickness in end-stage renal disease. Circulation
24. Feldman, H. I. et al. Iron administration and rHuEPO in hemodialysis patients. Am. J. Kidney 106, 2212–2217 (2002).
clinical outcomes in hemodialysis patients. Dis. 42, 325–330 (2003). 64. Himmelfarb, J. Uremic toxicity, oxidative stress,
J. Am. Soc. Nephrol. 13, 734–744 (2002). 44. Ofalabi, O., Velarde, C. & Besarab, A. Efficacy and and hemodialysis as renal replacement therapy.
25. Feldman, H. I. et al. Administration of parenteral tolerance of heme iron peptide (HIP) compared Semin. Dial. 22, 636–643 (2009).
iron and mortality among hemodialysis patients. to iron fumurate (IF) in CKD patients [abstract]. 65. Bishu, K. & Agarwal, R. Acute injury with
J. Am. Soc. Nephrol. 15, 1623–1632 (2004). J. Am. Soc. Nephrol. 14, 793A (2003). intravenous iron and concerns regarding long-
26. Nemeth, E. et al. Hepcidin regulates cellular iron 45. Sepandj, F., Jindal, K., West, M. & Hirsch, D. term safety. J. Am. Soc. Nephrol. 1 (Suppl. 1),
efflux by binding to ferroportin and inducing its Economic appraisal of maintenance parenteral S19–S23 (2006).
internalization. Science 306, 2090–2093 iron administration in treatment of the anaemia 66. Caussé, E. et al. Aminothiols and allantoin in
(2004). in chronic haemodialysis patients. Nephrol. Dial. chronic dialysis patients: effects of hemodialysis
27. Tanno, T., Noel, P. & Miller, J. L. Growth Transplant. 11, 319–322 (1996). sessions. Clin. Nephrol. 73, 51–57 (2010).
differentiation factor 15 in erythroid health and 46. Yee, J. & Besarab, A. Iron sucrose: the oldest 67. Shah, S. V. & Rajapurkar, M. M. The role of labile
disease. Curr. Opin. Hematol. 17, 184–190 therapy becomes new. Am. J. Kidney Dis. 40, iron in kidney disease and treatment with
(2010). 1111–1121 (2002). chelation. Hemoglobin 33, 378–385 (2009).
28. Chasis, J. A. & Mohandas, N. Erythroblastic 47. Bolaños, L. et al. Continuous intravenous sodium 68. Kalantar-Zadeh, K., Regidor, D. L.,
islands: niches for erythropoiesis. Blood 112, ferric gluconate improves efficacy in the McAllister, C. J., Michael, B. & Warnock, D. G.
470–478 (2008). maintenance phase of EPOrHu administration in Time-dependent associations between iron and
29. Babitt, J. L. & Lin, H. Y. Molecular mechanisms hemodialysis patients. Am. J. Nephrol. 22, 67–72 mortality in hemodialysis patients. J. Am. Soc.
of hepcidin regulation: implications for the (1992). Nephrol. 16, 3070–3080 (2005).
anemia of CKD. Am. J. Kidney Dis. 55, 726–741 48. Bregman, H. & Gelfand, M. C. Iron overload in 69. Kovesdy, C. S., Estrada, W., Abmahdzadeh, S. &
(2010). patients on maintenance hemodialysis. Int. J. Kalantar-Zadeh, K. association of iron markers of
30. Ganz, T., Olbina, G., Girelli, D., Nemeth, E. & Artif. Organs 4, 56–57 (1981). iron stores with outcomes in patients with
Westerman, M. Immunoassay for human serum 49. Ali, M. et al. Failure of serum ferritin levels to non‑dialysis‑dependent chronic kidney disease.
hepcidin. Blood 112, 4292–4297 (2008). predict bone-marrow iron content after Clin. J. Am. Soc. Nephrol. 4, 435–441 (2009).
31. Nemeth, E. Targeting the hepcidin–ferroportin intravenous iron-dextran therapy. Lancet 1, 70. Pollack, V. E., Lorxh, J. A., Schukla, R. &
axis in the diagnosis and treatment of anemias. 652–655 (1982). Satwah, S. The importance of iron in long-term
Adv. Hematol. 2010, 750643 (2010). 50. Gokal, R. et al. Iron metabolism in haemodialysis survival of maintenance hemodialysis patients
32. Fernández-Rodríguez, A. M. et al. Diagnosis of patients. A study of the management of iron treated with epoetin-alpha and intravenous iron:
iron deficiency in chronic renal failure. Am. J. therapy and overload. Q. J. Med. 48, 369–391 analysis of 9.5 years of prospectively collected
Kidney Dis. 34, 508–513 (1999). (1979). data. BMC Nephrol. 10, 6 (2009).
33. Kalantar-Zadeh, K. et al. Diagnosis of iron 51. Winchester, J. F. Management of iron overload in 71. Agarwal, R., Rizkala, A. R., Kaskas, M. O.,
deficiency anemia in renal failure patients during dialysis patients. Semin. Nephrol. 6, 22–26 Minasian, R. & Trout, J. R. Iron sucrose causes
the post-erythropoietin era. Am. J. Kidney Dis. (1986). greater proteinuria than ferric gluconate in non-
26, 292–299 (1995). 52. Bárány, P., Eriksson, L. C., Hultcrantz, R., dialysis chronic kidney disease. Kidney Int. 72,
34. Singh, A. K., Coyne, D. W., Shapiro, W., Pettersson, E. & Bergström, J. Serum ferritin and 638–642 (2007).
Rizkala, A. R.; DRIVE Study Group. Predictors of tissue iron in anemic dialysis patients. Miner. 72. Coyne, D. W. et al. Ferric gluconate is highly
the response to treatment in anemic Electrolyte Metab. 23, 273–276 (1997). efficacious in anemic hemodialysis patients with
hemodialysis patients with high serum ferritin 53. Rodgers, G. M. et al. High-molecular weight iron high serum ferritin and low transferrin saturation:
and low transferrin saturation. Kidney Int. 71, dextran: a wolf in sheep’s clothing? J. Am. Soc. results of the Dialysis Patients’ Response to IV
1163–1171 (2007). Nephrol. 19, 833–840 (2008). Iron with Elevated Ferritin (DRIVE) Study. J. Am.
35. Stancu, S. et al. Bone marrow iron, iron indices, 54. Raimundo, M., Abreu, F. & Da Costa, G. Soc. Nephrol. 18, 975–984 (2007).
and the response to intravenous iron in patients Parenteral iron safety in haemodialysis patients. 73. Anker, S. D. et al. Ferric carboxymaltose in
with non‑dialysis‑dependent CKD. Clin. J. Am. Port. J. Nephrol. Hyperten. 23, 153–159 (2009). patients with heart failure and iron deficiency.
Soc. Nephrol. 55, 639–647 (2010). 55. Nissenson, A. R., Lindsay, R. M., Swan, S., N. Engl. J. Med. 361, 2436–2448 (2009).
36. Stancu, S., Bârsan, L., Stanciu, A. & Seligman, P. & Strobos, J. Sodium ferric 74. Agarwal, R. A clinical trial of oral versus IV iron in
Mircescu, G. Can the response to iron therapy gluconate complex in surcrose is safe and patients with chronic kidney disease. ClinicalTrial.
be predicted in anemic nondialysis patients with effective in hemodialysis patients: gov [online], http://www.clinicaltrials.gov/ct2/
chronic kidney disease Clin. J. Am. Soc. Nephrol. North American Clinical Trial. Am. J. Kidney Dis. show/NCT00830037?term=NCT00830037&
5, 409–416 (2010). 33, 471–482 (1999). rank=1 (2010).
37. Bross, R. et al. Association of serum total iron- 56. Bailie, G. R., Johnson, C. A. & Mason, N. A. 75. Mircescu, G. Intravenous iron in patients with
binding capacity and its changes over time with Parenteral iron use in the management of severe chronic heart failure and chronic kidney
nutritional and clinical outcomes in hemodialysis anemia in end-stage renal disease patients. Am. disease. ClinicalTrial.gov [online], http://
patients. Am. J. Nephrol. 29, 571–581 (2009). J. Kidney Dis. 35, 1–12 (2000). www.clinicaltrials.gov/ct2/show/NCT00384657
38. Kalantar-Zadeh, K., McAllister, C. J., Lehn, R. S., 57. Chandler, G., Harchawal, K. & Macdougall, I. C. ?term=NCT00384657&rank=1 (2010).
Liu, E. & Kopple, J. D. A low serum iron level is a Intravenous iron sucrose: establishing a safe 76. Dahl, N. V., Henry, D. H. & Coyne, D. W.
predictor of poor outcome in hemodialysis dose. Am. J. Kidney Dis. 38, 988–991 (2001). Thrombosis with erythropoietic stimulating
patients. Am. J. Kidney Dis. 43, 671–684 58. Spinowitz, B. S. et al. Ferumoxytol for treating agents—does iron-deficient erythropoiesis play a
(2004). iron deficiency anemia in CKD. J. Am. Soc. role? Semin. Dial. 21, 210–211 (2008).
39. Vaghefi, N. et al. Effect of stabilising amino acids Nephrol. 19, 1599–1605 (2008). 77. Streja, E. et al. Erythropoietin, iron depletion, and
on the digestive absorption of heme and non- 59. Singh, A. et al. Safety of ferumoxytol in patients relative thrombocytosis: a possible explanation
heme iron. Reprod. Nutr. Dev. 38, 559–566 with anemia and, CKD. Am. J. Kidney Dis. 52, for hemoglobin–survival paradox in hemodialysis.
(1998). 907–915 (2008). Am. J. Kidney Dis. 52, 727–736 (2008).

NATURE REVIEWS | NEPHROLOGY VOLUME 6  |  DECEMBER 2010  |  709


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

78. Besarab, A., Rayamajhi, S., Al-Sharif, H., 83. Soman, S., Zasuwa, G. & Yee, J. Automation, administration. Nephrol. Dial. Transplant.
Frinak, S. & Yee, J. Iron repletion decreases decision support, and expert systems in 10 (Suppl. 6), 44–47 (1995).
platelet counts (PLT) in non-dialysis CKD nephrology. Adv. Chronic Kidney Dis. 15, 42–55 88. Attallah, N., Osman-Malik, Y., Frinak, S. &
patients [abstract 50]. Am. J. Kidney Dis. 55 (2008). Besarab, A. Effect of intravenous ascorbic
(Suppl.), B54 (2010). 84. Covic, A. & Mircescu, G. The safety and acid in hemodialysis patients with
79. Vaziri, N. D. Thrombocytosis in EPO–treated efficacy of intravenous ferric carboxymaltose erythropoietin-hyporesponsive anemia and
dialysis patients may be mediated by EPO rather in anaemic patients undergoing haemodialysis: hyperferritinemia. Am. J. Kidney Dis. 47,
than iron deficiency. Am. J. Kidney Dis. 53, a multi-centre, open-label, clinical study. 644–654 (2006).
733–736 (2009). Nephrol. Dial. Transplant. 25, 2722–2730 89. Goch, J., Birgegård, G., Danielson, B. G. &
80. Gotloib, L., Silverberg, D., Fudin, R. & Shostak, A. (2010). Wikström, B. Treatment of erythropoietin-
Iron deficiency is a common cause of anemia in 85. Provenzano, R. et al. Ferumoxytol as an resistant anaemia with desferrioxamine in
chronic kidney disease and can often be intravenous iron replacement therapy in patients on haemofiltration. Eur. J. Haematol. 55,
corrected with intravenous iron. J. Nephrol. 19, hemodialysis patients. Clin. J. Am. Soc. Nephrol. 73–77 (1995).
161–167 (2006). 4, 386–393 (2009). 90. Sargent, J. A. & Acchiardo, S. R. Iron
81. Macdougall, I. C. Iron supplementation in the 86. Gupta, A. et al. Dialysate iron therapy: infusion of requirements in hemodialysis. Blood Purif. 2,
non-dialysis chronic kidney disease (ND-CKD) soluble ferric pyprophosphate via the dialysate 12–23 (2004).
patient: oral or intravenous? Curr. Med. Res. during hemodialysis. Kidney Int. 55, 1891–1898
Opin. 26, 473–482 (2010). (1999).
82. Pfeffer, M. A. et al. A trial of darbepoetin alfa in 87. Gastaldello, K. et al. Resistance to Author contributions
type 2 diabetes and chronic kidney disease. erythropoietin in iron-overloaded haemodialysis A. Besarab and D. W. Coyne contributed equally to all
N. Engl. J. Med. 361, 2019–2032 (2009). patients can be overcome by ascorbic acid aspects of this manuscript.

710  |  DECEMBER 2010  |  VOLUME 6  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

CORRECTION
Iron supplementation to treat anemia in patients with chronic
kidney disease
Anatole Besarab & Daniel W. Coyne
Nat. Rev. Nephrol. 6, 699–710; doi:10.1038/nrneph.2010.139
In the version of this article initially published online and in print, incorrect volume and
page numbers were given for reference 90. The correct reference is Sargent, J. A. &
Acchiardo, S. R. Iron requirements in hemodialysis. Blood Purif. 22, 112–123 (2004).
The error has been corrected for the HTML and PDF versions of the article.

NATURE REVIEWS | NEPHROLOGY


© 2011 Macmillan Publishers Limited. All rights reserved

You might also like