Sie sind auf Seite 1von 14

The new england journal of medicine

review article

medical progress

Anemia of Chronic Disease


Guenter Weiss, M.D., and Lawrence T. Goodnough, M.D.

a nemia of chronic disease, the anemia that is the second most


prevalent after anemia caused by iron deficiency, occurs in patients with acute
or chronic immune activation.1-4 The condition has thus been termed anemia
of inflammation.1-4 The most frequent conditions associated with anemia of chronic
disease are listed in Table 1.5-22
From the Department of General Internal
Medicine, Clinical Immunology and Infec-
tious Diseases, Medical University of Inns-
bruck, Innsbruck, Austria (G.W.); and the
Departments of Pathology and Medicine,
Stanford University, Stanford, Calif. (L.T.G.).
Address reprint requests to Dr. Weiss at
the Department of General Internal Medi-
pathophysiological features cine, Clinical Immunology and Infectious
Diseases, Medical University of Innsbruck,
Anichstr. 35, A-6020 Innsbruck, Austria, or
Anemia of chronic disease is immune driven; cytokines and cells of the reticuloendo- at guenter.weiss@uibk.ac.at.
thelial system induce changes in iron homeostasis, the proliferation of erythroid pro-
genitor cells, the production of erythropoietin, and the life span of red cells, all of which N Engl J Med 2005;352:1011-23.
Copyright 2005 Massachusetts Medical Society.
contribute to the pathogenesis of anemia (Fig. 1). Erythropoiesis can be affected by dis-
ease underlying anemia of chronic disease through the infiltration of tumor cells into
bone marrow or of microorganisms, as seen in human immunodeficiency virus (HIV)
infection, hepatitis C, and malaria.23,24 Moreover, tumor cells can produce proinflam-
matory cytokines and free radicals that damage erythroid progenitor cells.3,4,8 Bleeding
episodes, vitamin deficiencies (e.g., of cobalamin and folic acid), hypersplenism, auto-
immune hemolysis, renal dysfunction, and radio- and chemotherapeutic interventions
themselves can also aggravate anemia.25,26
Anemia with chronic kidney disease shares some of the characteristics of anemia of
chronic disease, although the decrease in the production of erythropoietin, mediated by
renal insufficiency and the antiproliferative effects of accumulating uremic toxins, con-
tribute importantly.27 In addition, in patients with end-stage renal disease, chronic im-
mune activation can arise from contact activation of immune cells by dialysis mem-
branes, from frequent episodes of infection, or from both factors, and such patients
present with changes in the homeostasis of body iron that is typical of anemia of
chronic disease.27

dysregulation of iron homeostasis


A hallmark of anemia of chronic disease is the development of disturbances of iron
homeostasis, with increased uptake and retention of iron within cells of the reticuloen-
dothelial system. This leads to a diversion of iron from the circulation into storage sites
of the reticuloendothelial system, subsequent limitation of the availability of iron for
erythroid progenitor cells, and iron-restricted erythropoiesis.
In mice that are injected with the proinflammatory cytokines interleukin-1 and tumor
necrosis factor a (TNF-a), both hypoferremia and anemia develop28; this combination
of conditions has been linked to cytokine-inducible synthesis of ferritin, the major pro-
tein associated with iron storage, by macrophages and hepatocytes.29 In chronic inflam-
mation, the acquisition of iron by macrophages most prominently takes place through
erythrophagocytosis30 and the transmembrane import of ferrous iron by the protein di-
valent metal transporter 1 (DMT1).31

n engl j med 352;10 www.nejm.org march 10, 2005 1011


The new england journal of medicine

Table 1. Underlying Causes of Anemia of Chronic Figure 1 (facing page). Pathophysiological Mechanisms
Disease. Underlying Anemia of Chronic Disease.
Estimated In Panel A, the invasion of microorganisms, the emer-
Associated Diseases Prevalence* gence of malignant cells, or autoimmune dysregulation
leads to activation of T cells (CD3+) and monocytes.
percent These cells induce immune effector mechanisms, there-
Infections (acute and chronic) 18958-10 by producing cytokines such as interferon-g (from T cells)
and tumor necrosis factor a (TNF-a), interleukin-1, in-
Viral infections, including human
immunodeficiency virus infection terleukin-6, and interleukin-10 (from monocytes or mac-
rophages). In Panel B, interleukin-6 and lipopolysaccha-
Bacterial
ride stimulate the hepatic expression of the acute-phase
Parasitic protein hepcidin, which inhibits duodenal absorption of
Fungal iron. In Panel C, interferon-g, lipopolysaccharide, or both
increase the expression of divalent metal transporter 1
Cancer 30779,12-14
on macrophages and stimulate the uptake of ferrous iron
Hematologic (Fe2+). The antiinflammatory cytokine interleukin-10 up-
Solid tumor regulates transferrin receptor expression and increases
Autoimmune 8715,9,15,16 transferrin-receptormediated uptake of transferrin-
bound iron into monocytes. In addition, activated mac-
Rheumatoid arthritis
rophages phagocytose and degrade senescent erythro-
Systemic lupus erythematosus cytes for the recycling of iron, a process that is further in-
and connective-tissue diseases duced by TNF-a through damaging of erythrocyte
Vasculitis membranes and stimulation of phagocytosis. Interferon-g
Sarcoidosis and lipopolysaccharide down-regulate the expression of
the macrophage iron transporter ferroportin 1, thus in-
Inflammatory bowel disease hibiting iron export from macrophages, a process that is
Chronic rejection after solid-organ trans- 87017-19 also affected by hepcidin. At the same time, TNF-a, inter-
plantation leukin-1, interleukin-6, and interleukin-10 induce ferritin
Chronic kidney disease and inflammation 235020-22 expression and stimulate the storage and retention of
iron within macrophages. In summary, these mecha-
* Values shown are ranges. Epidemiologic data are not nisms lead to a decreased iron concentration in the cir-
available for all conditions associated with the anemia of culation and thus to a limited availability of iron for eryth-
chronic disease. The prevalence and severity of anemia roid cells. In Panel D, TNF-a and interferon-g inhibit the
are correlated with the stage of the underlying condition5,6 production of erythropoietin in the kidney. In Panel E,
and appear to increase with advanced age.7 TNF-a, interferon-g, and interleukin-1 directly inhibit the
The prevalence of anemia in patients with cancer is af- differentiation and proliferation of erythroid progenitor
fected by therapeutic procedures and age. A high preva- cells. In addition, the limited availability of iron and the
lence was reported in one study in which 77 percent of el-
decreased biologic activity of erythropoietin lead to inhi-
derly men and 68 percent of elderly women with cancer
were anemic.11 In another study, anemia was observed in bition of erythropoiesis and the development of anemia.
41 percent of patients with solid tumors before radio- Plus signs represent stimulation, and minus signs inhi-
therapy and in 54 percent thereafter.12 bition.

Interferon-g, lipopolysaccharide, and TNF-a up- macrophages and by translational stimulation of


regulate the expression of DMT1, with an increased ferritin expression34 (Fig. 1).
uptake of iron into activated macrophages.32 These The identification of hepcidin, an iron-regulated
proinflammatory stimuli also induce the retention acute-phase protein that is composed of 25 amino
of iron in macrophages by down-regulating the ex- acids, helped to shed light on the relationship of the
pression of ferroportin, thus blocking the release of immune response to iron homeostasis and anemia
iron from these cells.32 Ferroportin is a transmem- of chronic disease. Hepcidin expression is induced
brane exporter of iron, a process that is believed to by lipopolysaccharide and interleukin-6 and is in-
be responsible for the transfer of absorbed ferrous hibited by TNF-a.35 Transgenic or constitutive over-
iron from duodenal enterocytes to the circulation.33 expression of hepcidin results in severe iron-defi-
Moreover, antiinflammatory cytokines such as inter- ciency anemia in mice.36 Inflammation in mice that
leukin-10 can induce anemia through the stimula- are hepcidin-deficient did not lead to hypoferremia,
tion of transferrin-mediated acquisition of iron by a finding that suggests that hepcidin may be central-

1012 n engl j med 352;10 www.nejm.org march 10 , 2005


medical progress

n engl j med 352;10 www.nejm.org march 10, 2005 1013


The new england journal of medicine

ly involved in the diversion of iron traffic through chronic disease are inadequate for the degree of
decreased duodenal absorption of iron and the anemia in most, but not all, conditions.43,44 The
blocking of iron release from macrophages that oc- cytokines interleukin-1 and TNF-a directly inhibit
curs in anemia of chronic disease (Fig. 1).35,37 The erythropoietin expression in vitro45 a finding
induction of hypoferremia by interleukin-6 and hep- that is probably due, at least in part, to cytokine-me-
cidin occurs within a few hours and is not observed diated formation of reactive oxygen species, which
in interleukin-6knockout mice that are treated with in turn affects the binding affinities of erythropoi-
turpentine as a model of inflammation, a finding etin-inducing transcription factors and also dam-
that suggests that hepcidin may be central to ane- ages erythropoietin-producing cells. Although
mia of chronic disease.38 A recently identified gene, convincing data from human studies are lacking,
hemojuvelin, may act in concert with hepcidin in the injection of lipopolysaccharide into mice re-
inducing these changes.39 Accordingly, the distur- sults in reduced expression of erythropoietin
bance of iron homeostasis with subsequent limita- mRNA in kidneys and decreased levels of circulat-
tion of the availability of iron for erythroid progen- ing erythropoietin.45
itor cells appears to impair the proliferation of The responsiveness of erythroid progenitor cells
these cells by negatively affecting heme biosynthe- to erythropoietin appears to be inversely related to
sis (Table 2). the severity of the underlying chronic disease and
the amount of circulating cytokines, since in the
impaired proliferation of erythroid presence of high concentrations of interferon-g or
progenitor cells TNF-a, much higher amounts of erythropoietin are
In patients with anemia of chronic disease, the pro- required to restore the formation of erythroid col-
liferation and differentiation of erythroid precursors ony-forming units.46 After binding to its receptor,
erythroid burst-forming units and erythroid col- erythropoietin stimulates members of the signal
ony-forming units are impaired4 and are linked transduction pathways and subsequently activates
to the inhibitory effects of interferon-a, -b, and -g, mitogen and tyrosine kinase phosphorylation, pro-
TNF-a, and interleukin-1, which influence the cesses affected by the inflammatory cytokines and
growth of erythroid burst-forming units and eryth- the negative-feedback regulation they induce.45,47
roid colony-forming units.4 Interferon-g appears to The response to erythropoietin is further reduced
be the most potent inhibitor,40 as reflected by its in- by the inhibitory effects of proinflammatory cyto-
verse correlation with hemoglobin concentrations kines toward the proliferation of erythroid progen-
and reticulocyte counts.6 The underlying mecha- itor cells, the parallel down-regulation of erythro-
nisms may involve cytokine-mediated induction of poietin receptors, and the limited availability of iron
apoptosis, which appears, in part, related to the for- to contribute to cell proliferation and hemoglobin
mation of ceramide, the down-regulation of the ex- synthesis. Finally, increased erythrophagocytosis
pression of erythropoietin receptors on progenitor during inflammation leads to a decreased erythro-
cells, impaired formation and activity of erythro- cyte half-life, along with anticipated damage to
poietin, and a reduced expression of other pro- erythrocytes that is mediated by cytokines and free
hematopoietic factors, such as stem-cell fac- radicals (Table 2).48,49
tor.4,40,41 Moreover, cytokines exert direct toxic
effects on progenitor cells by inducing the forma- laboratory evaluation
tion of labile free radicals such as nitric oxide or su-
peroxide anion by neighboring macrophage-like iron status
cells (Table 2).42 Anemia of chronic disease is a normochromic, nor-
mocytic anemia that is characteristically mild (he-
blunted erythropoietin response moglobin level, 9.5 g per deciliter) to moderate
Erythropoietin regulates erythroid-cell proliferation (hemoglobin level, 8 g per deciliter). Patients with
centrally. Erythropoietin expression is inversely re- the condition have a low reticulocyte count, which
lated to tissue oxygenation and hemoglobin levels, indicates underproduction of red cells. A definitive
and there is a semilogarithmic relation between the diagnosis may be hampered by coexisting blood
erythropoietin response (log) and the degree of ane- loss, the effects of medications, or inborn errors of
mia (linear). Erythropoietin responses in anemia of hemoglobin synthesis such as thalassemia. The

1014 n engl j med 352;10 www.nejm.org march 10 , 2005


medical progress

Table 2. Pathophysiological Factors in Anemia of Chronic Disease.*


Feature Key Factors Mechanisms Cellular Pathway Systemic Effects
Pathologic TNF-a or inter- Induces ferritin transcription Increased iron storage within the RES Hypoferremia, hyperfer-
iron homeo- leukin-1 Leads to a decreased erythrocyte ritinemia
stasis half-life, mediated by TNF-a Unknown (may be through damage of Erythrophagocytosis
red cells by radicals)
Interleukin-6 Induces ferritin transcription Increased iron storage within the RES Hypoferremia, hyperfer-
or translation ritinemia
Stimulates formation of hepcidin Iron absorption and export from macrophag- Hypoferremia
es decreased by hepcidin
Interferon-g or Stimulates DMT1 synthesis; Increased iron uptake and inhibition of iron Hypoferremia
lipopolysac- down-regulates ferroportin 1 recirculation (e.g., derived from erythro-
charide expression phagocytosis) in macrophages
Interleukin-10 Induces transferrin-receptor ex- Increased uptake and storage of transferrin- Hypoferremia, hyperfer-
pression; stimulates ferritin bound iron in macrophages ritinemia
translation
Erythrophagocy- Reduces erythrocyte half-life Recirculated iron restricted within macro- Hypoferremia, anemia
tosis through increased uptake phages
of erythrocytes damaged by
TNF-a
Impaired Interferon-g, in- Inhibits proliferation and differen- Induction of apoptosis; down-regulation Anemia with normal or
erythro- terleukin-1, tiation of CFU-E and BFU-E of erythropoietin-receptor expression; decreased reticulocyte
poiesis or TNF-a reduced formation of stem-cell factor counts
Causes hypoferremia through Iron-restricted erythropoiesis Anemia with increased
diversion of iron to the RES levels of tin protopor-
phyrin
Induces formation of nitric oxide Erythroid aminolevulinate synthase inhibited Anemia with increased
levels of levulinic acid
Alpha1- Limits iron uptake by erythroid Reduced proliferation of BFU-E or CFU-E Anemia
antitrypsin cells
Tumor cells or Infiltrate bone marrow Displacement of progenitor cells Anemia, pancytopenia,
microbes or both
Produce soluble mediators Local inflammation and formation of cyto- Anemia, pancytopenia,
kines and radicals or both
Consume vitamins Inhibition of progenitor-cell proliferation Systemic deficiency of
folate or cobalamin
Hypoferremia Caused by cytokine-mediated di- Impaired heme biosynthesis, erythropoietin Anemia
version of iron into the RES responsiveness; reduced proliferation
and reduced iron absorption of CFU-E
Blunted eryth- Erythropoietin Inhibits erythropoietin produc- Reduction of erythropoietin transcription; Decreased levels of circu-
ropoietin deficiency tion by interleukin-1 and radical-mediated damage of erythropoi- lating erythropoietin
response TNF-a etin-producing cells
Hypoferremia Reduces erythropoietin respon- Blunted heme biosynthesis and progenitor- Anemia, hypoferremia
siveness of progenitor cells cell proliferation
owing to iron restriction
Interferon-g, in- Impair responsiveness of progeni- Reduced erythropoietin-receptor expression Anemia
terleukin-1, tor cells to erythropoietin on CFU-E; damage of erythroid progeni-
and TNF-a tors mediated by cytokines or radicals;
possible interference with erythropoietin
signal transduction

* TNF-a denotes tumor necrosis factor a, RES reticuloendothelial system, DMT1 divalent metal transporter 1, CFU-E erythroid colony-forming
units, and BFU-E erythroid burst-forming units.

evaluation of anemia of chronic disease must also ciency anemia thus relates to the latter as an abso-
include a determination of the status of whole-body lute iron deficiency, whereas the pathophysiology
iron in order to rule out iron-deficiency anemia,2-4,49 of anemia of chronic disease is multifactorial, as de-
usually hypochromic and microcytic. The difference scribed in Table 2.
between anemia of chronic disease and iron-defi- In both anemia of chronic disease and iron-

n engl j med 352;10 www.nejm.org march 10, 2005 1015


The new england journal of medicine

deficiency anemia, the serum concentration of iron within the reticuloendothelial system, along with in-
and transferrin saturation are reduced, reflecting creased ferritin levels due to immune activation.29
absolute iron deficiency in iron-deficiency anemia The soluble transferrin receptor is a truncated
and hypoferremia due to acquisition of iron by the fragment of the membrane receptor that is in-
reticuloendothelial system in anemia of chronic dis- creased in iron deficiency, when the availability of
ease.2-4 In the case of anemia of chronic disease, iron for erythropoiesis is low.52 In contrast, levels of
the decrease in transferrin saturation is primarily a soluble transferrin receptors in anemia of chronic
reflection of decreased levels of serum iron. In iron- disease are not significantly different from normal,
deficiency anemia, transferrin saturation may be because transferrin-receptor expression is negative-
even lower because serum concentrations of the ly affected by inflammatory cytokines.53 A determi-
iron transporter transferrin are increased, whereas nation of the levels of soluble transferrin receptors
transferrin levels remain normal or are decreased by means of commercially available assays can be
in anemia of chronic disease. helpful for differentiation between patients with
The search for an underlying cause of iron defi- anemia of chronic disease alone (with either nor-
ciency should include a history taking to rule out a mal or high ferritin levels and low levels of soluble
dietary cause.42 Frequently, iron deficiency indicates transferrin receptors) and patients with anemia of
pathological blood loss such as an increased loss chronic disease with accompanying iron deficiency
of menstrual blood in women or chronic gastroin- (with low ferritin levels and high levels of soluble
testinal bleeding in the setting of ulcerative gastro- transferrin receptors).52,54
intestinal disease, inflammatory bowel disease, As compared with patients who have anemia
angiodysplasia, colon adenomas, gastrointestinal of chronic disease alone, patients with anemia of
cancer, or parasitic infections. chronic disease and concomitant iron-deficiency
Ferritin is used as a marker of iron storage, and anemia more frequently have microcytes, and their
a level of 15 ng per milliliter is generally taken as anemia tends to be more severe. The ratio of the
indicating absent iron stores.50 However, a ferritin concentration of soluble transferrin receptors to the
level of 30 ng per milliliter provides better positive log of the ferritin level may also be helpful.52 A ra-
predictive values for iron-deficiency anemia (92 to tio of less than 1 suggests anemia of chronic dis-
98 percent) when studied in several populations.51 ease, whereas a ratio of more than 2 suggests ab-
For patients with anemia of chronic disease, how- solute iron deficiency coexisting with anemia of
ever, ferritin levels are normal or increased (Table 3), chronic disease (Table 3). The determination of the
reflecting increased storage and retention of iron percentage of hypochromic red cells or reticulocyte
hemoglobin content can also be useful in detecting
accompanying iron-restricted erythropoiesis in pa-
Table 3. Serum Levels That Differentiate Anemia of Chronic Disease tients with anemia of chronic disease.54
from Iron-Deficiency Anemia.*
erythropoietin
Anemia of Iron-Deficiency Both
Variable Chronic Disease Anemia Conditions Measurement of erythropoietin levels is useful only
for anemic patients with hemoglobin levels of less
Iron Reduced Reduced Reduced
than 10 g per deciliter, since erythropoietin levels at
Transferrin Reduced Increased Reduced
to normal higher hemoglobin concentrations remain well in
Transferrin saturation Reduced Reduced Reduced
the normal range.43 Furthermore, any interpreta-
tion of an erythropoietin level in anemia of chronic
Ferritin Normal Reduced Reduced
to increased to normal disease with a hemoglobin level less than 10 g per
Soluble transferrin receptor Normal Increased Normal deciliter must take into account the degree of ane-
to increased mia.55,56 Erythropoietin levels have been analyzed
Ratio of soluble transferrin Low (<1) High (>2) High (>2) for their predictive value with respect to the response
receptor to log ferritin to treatment of anemia of chronic disease with eryth-
Cytokine levels Increased Normal Increased ropoietic agents.13,57 After treatment with recombi-
nant human erythropoietin (epoetin) for two weeks,
* Relative changes are given in relation to the respective normal values. either a serum erythropoietin level of more than
Patients with both conditions include those with anemia of chronic disease
and true iron deficiency. 100 U per liter or a ferritin level of more than 400
ng per milliliter predicts a lack of response in 88

1016 n engl j med 352;10 www.nejm.org march 10 , 2005


medical progress

percent of patients with cancer who are not receiv- be optimal. A prospective, multicenter trial involv-
ing concomitant chemotherapy.13 Such predictors ing patients who are undergoing dialysis a study
have not been validated in patients with cancer who of an intervention that is designed to achieve nor-
are undergoing chemotherapy.57 Rather, changes in mal hematocrit levels (above 42 percent), as com-
hemoglobin levels or reticulocyte counts over time pared with lower levels (above 30 percent), with the
indicate a response to treatment with epoetin.57 use of a combination of erythropoietin therapy and
intravenous iron dextran was halted because of
treatment increased mortality in the high-hematocrit cohort.64
The patients who had high hematocrit levels in that
rationale for treatment study received higher doses of erythropoietin and
The rationale for the treatment of anemia of chron- intravenous iron than did patients who had low he-
ic disease is based on two principles. First, anemia matocrit levels. The link between iron stores and
can be generally deleterious in itself, requiring a morbidity or mortality rates is controversial, since
compensatory increase in cardiac output in order to it involves issues that are related to infections in pa-
maintain systemic oxygen delivery; second, anemia tients undergoing dialysis65 and detrimental coro-
is associated with a poorer prognosis in a variety of nary outcomes in men.66 An editorial67 concluded
conditions. Thus, moderate anemia warrants cor- that intravenous iron should be administered, if
rection, especially in patients older than 65 years of necessary, to improve the response to therapy with
age, those with additional risk factors (such as cor- epoetin in order to reach a target hematocrit of 33 to
onary artery disease, pulmonary disease, or chron- 36 percent in patients with chronic kidney disease.62
ic kidney disease), or a combination of these fac- Careful studies of the potentially harmful effects of
tors.7,58 In patients with renal failure who are iron supplementation in patients with various forms
receiving dialysis and in patients with cancer who of anemia of chronic disease are still needed.
are undergoing chemotherapy, correction of anemia Despite management guidelines, anemia of
up to hemoglobin levels of 12 g per deciliter is asso- chronic disease remains underrecognized and un-
ciated with an improvement in the quality of life.59,60 dertreated. In a study of 200,000 patients enrolled
Anemia has been associated with a relatively poor in a health maintenance organization between 1994
prognosis among patients with various conditions, and 1997, 23 percent of patients with chronic kid-
including cancer, chronic kidney disease, and con- ney disease had hematocrit levels under 30 percent,
gestive heart failure.9 This relationship has been ex- and only 30 percent of those with hematocrit levels
plored most fully in patients undergoing long-term below the target were receiving treatment for ane-
hemodialysis. In a retrospective review of nearly mia.9 It is important to note that anemia of chronic
100,000 patients undergoing hemodialysis, levels disease, if marked, can be a reflection of a more pro-
of hemoglobin of 8 g per deciliter or less were asso- gressive underlying disease.3,4,6,49 Thus, the notion
ciated with a doubling of the odds of death, as com- that correction of anemia alone may improve the
pared with hemoglobin levels of 10 to 11 g per deci- prognosis of other underlying chronic diseases
liter.61 Moreover, the odds ratios for death among such as cancer or inflammatory disease remains un-
patients who entered the study with hematocrit lev- proven.
els that were under 30 percent but that increased to
30 percent or more did not differ from those of pa- treatment options
tients who began and finished the study with he- When possible, treatment of the underlying disease
matocrit levels of 30 percent or more. Subsequent is the therapeutic approach of choice for anemia of
analyses have determined that hematocrit levels that chronic disease.3-5 Improvement in hemoglobin
were maintained between 33 and 36 percent were levels has been demonstrated, for example, in pa-
associated with the lowest risk of death among pa- tients with rheumatoid disease68 who were receiv-
tients undergoing dialysis.20,21 This evidence con- ing therapy with anti-TNF antibodies. In cases in
tributed to the development of guidelines for the which treating the underlying disease is not feasi-
management of anemia in patients with cancer or ble, alternative strategies are necessary (Table 4).
chronic kidney disease, guidelines that recommend
a target hemoglobin level of 11 to 12 g per deci- Transfusion
liter.14,62,63 Blood transfusions are widely used as a rapid and
However, a normal target hematocrit may not effective therapeutic intervention. Transfusions are

n engl j med 352;10 www.nejm.org march 10, 2005 1017


The new england journal of medicine

patients with anemia of chronic disease is contro-


Table 4. Therapeutic Options for the Treatment of Patients with Anemia
versial.72 Iron is an essential nutrient for prolifer-
of Chronic Disease.
ating microorganisms, and the sequestration of iron
Anemia of Chronic Anemia of Chronic Disease from microorganisms or tumor cells into the retic-
Treatment Disease with True Iron Deficiency uloendothelial system is believed to be a potential-
Treatment of underlying Yes Yes ly effective defense strategy to inhibit the growth of
disease pathogens.72 A study investigating measures to
Transfusions* Yes Yes predict the risk of bacteremia among patients
Iron supplementation No Yes undergoing hemodialysis who are receiving iron
Erythropoietic agents Yes Yes, in patients who do not have
parenterally showed that patients with a transfer-
a response to iron therapy rin saturation above 20 percent and ferritin levels
greater than 100 ng per milliliter had a significantly
* This treatment is for the short-term correction of severe or life-threatening higher risk of developing bacteremia,73 possibly at
anemia. Potentially adverse immunomodulatory effects of blood transfusions least in part because of the fact that iron has an in-
are controversial.
Although iron therapy is indicated for the correction of anemia of chronic dis- hibitory effect on cellular immune function that can
ease in association with absolute iron deficiency, no data from prospective be traced back to down-regulation of interferon-
studies are available on the effects of iron therapy on the course of underlying gmediated immune effector pathways.53 In addi-
chronic disease.
Overcorrection of anemia (hemoglobin >12 g per deciliter) may be potentially tion, iron therapy in a setting of long-term immune
harmful to patients; the clinical significance of erythropoietin-receptor expres- activation promotes the formation of highly toxic
sion on certain tumor cells needs to be investigated. hydroxyl radicals that can cause tissue damage and
endothelial dysfunction and increase the risk of
acute cardiovascular events.65,66,72
particularly helpful in the context of either severe On the other hand, iron therapy may confer ben-
anemia (in which the hemoglobin is less than 8.0 g efit. By inhibiting the formation of TNF-a, iron ther-
per deciliter) or life-threatening anemia (in which apy may reduce disease activity in rheumatoid ar-
the hemoglobin is less than 6.5 g per deciliter), par- thritis or end-stage renal disease.74,75 Furthermore,
ticularly when the condition is aggravated by com- patients with inflammatory bowel disease and ane-
plications that involve bleeding. Blood-transfusion mia respond well to parenteral iron therapy, with an
therapy has been associated with increased survival increase in hemoglobin levels.15
rates in anemic patients with myocardial infarc- In addition to possible absolute iron deficiency
tion,69 but transfusion itself has also been associ- accompanying the anemia of chronic disease, func-
ated with multiorgan failure and increased mortal- tional iron deficiency develops under conditions
ity in patients who are in critical care.70 Whether of intense erythropoiesis54,76 during therapy with
blood transfusions modulate the immune system, erythropoietic agents, with a decrease in transferrin
causing clinically relevant adverse effects, remains saturation and ferritin to levels 50 to 75 percent be-
undetermined.71 It is important to note that exist- low baseline.54,76 Parenteral iron has been demon-
ing guidelines for the management of anemia of strated to enhance rates of response to therapy with
chronic disease in patients with cancer or chronic erythropoietic agents in patients with cancer who
kidney disease do not recommend long-term blood are undergoing chemotherapy77 and in patients un-
transfusion therapy in their management algo- dergoing dialysis.62
rithms because of the risks associated with long- On the basis of current data,77 patients with ane-
term transfusion, such as iron overload and sensi- mia of chronic disease and absolute iron deficiency
tization to HLA antigens that may occur in patients should receive supplemental iron therapy.14,62,63
before renal transplantation.53-55 Iron supplementation should also be considered for
patients who are unresponsive to therapy with eryth-
Iron Therapy ropoietic agents because of functional iron deficien-
Oral iron is poorly absorbed because of the down- cy. In this setting, iron is more likely to be absorbed
regulation of absorption in the duodenum.37,38 Only and utilized by the erythron rather than by patho-
a fraction of the absorbed iron will reach the sites gens, as indicated by an increase in hemoglobin
of erythropoiesis, owing to iron diversion mediat- levels without demonstrable infectious complica-
ed by cytokines, which directs iron into the reticu- tions.73,77 However, iron therapy is currently not
loendothelial system. In addition, iron therapy for recommended for patients with anemia of chronic

1018 n engl j med 352;10 www.nejm.org march 10, 2005


medical progress

disease who have a high or normal ferritin level logic effects, including interference with the signal-
(above 100 ng per milliliter), owing to possible ad- transduction cascade of cytokines.45 For example,
verse outcomes in this setting.53,65,66,72,78,79 the long-term administration of epoetin has been
reported to decrease levels of TNF-a in patients with
Erythropoietic Agents chronic kidney disease81; reportedly, those who re-
Erythropoietic agents for patients with anemia of sponded well to epoetin therapy had a significantly
chronic disease are currently approved for use by higher level of expression of CD28 on T cells and
patients with cancer who are undergoing chemo- lower levels of interleukin-10, interleukin-12, in-
therapy, patients with chronic kidney disease, and terferon-g, and TNF-a than did those with a poor
patients with HIV infection who are undergoing response.85 Such antiinflammatory effects might
myelosuppressive therapy. The percentage of pa- be of benefit in certain diseases such as rheumatoid
tients with anemia of chronic disease who respond arthritis, a disease in which combined treatment
to therapy with erythropoietic agents is 25 percent in with epoetin and iron not only increased hemoglo-
myelodysplastic syndromes,80 80 percent in multi- bin levels but also resulted in a reduction of disease
ple myeloma,81 and up to 95 percent in rheumatoid activity.74
arthritis and chronic kidney disease.62 The thera- In addition, erythropoietin receptors are found
peutic effect involves counteracting the antiprolif- on several malignant cell lines, including mamma-
erative effects of cytokines,46,49 along with the stim- ry, ovarian, uterine, prostate, hepatocellular, and re-
ulation of iron uptake and heme biosynthesis in nal carcinomas, as well as on myeloid cell lines.86-88
erythroid progenitor cells.3 Accordingly, a poor re- However, there are contradictory reports concern-
sponse to treatment with erythropoietic agents is ing the effects of treatment with epoetin on such
associated with increased levels of proinflammato- cells. Although the drug led to tumor regression in
ry cytokines, on the one hand, and poor iron avail- a murine model of myeloma,89 administration to
ability, on the other hand.13,76,82 erythropoietin-receptorexpressing human renal-
Three erythropoietic agents are currently avail- carcinoma cells in vitro stimulated their prolifera-
able epoetin alfa, epoetin beta, and darbepoetin tion.90 High amounts of erythropoietin receptors
alfa, which differ in terms of their pharmacologic are found in 90 percent of biopsies from human
compounding modifications, receptor-binding af- breast carcinomas.91 The production of erythropoi-
finity, and serum half-life, thus allowing for alter- etin receptors by cancer cells appears to be regulat-
native dosing and scheduling strategies.83 Concern ed by hypoxia, and in clinical cancer specimens the
was recently aroused by the identification of 191 highest levels of erythropoietin receptors were as-
epoetin-associated cases of pure red-cell aplasia be- sociated with neoangiogenesis, tumor hypoxia, and
tween 1998 and 2004, as compared with only 3 such infiltrating tumors.88,91 Potentially adverse effects
cases between 1988 and 1998.84 The estimated ex- may be due to induction of neoangiogenesis by the
posure-adjusted incidence was 18 cases per 100,000 hormone, since erythropoietin increases inflamma-
patient-years for the formulation of epoetin alfa in tion and ischemia-induced neovascularization by
Eprex (Janssen-Cilag) without human serum albu- enhancing the mobilization of endothelial pro-
min, 6 cases per 100,000 patient-years for the Eprex genitor cells.92 Implantation of erythropoietin-
formulation with serum albumin, 1 case per 100,000 receptorexpressing cell lines into nude mice with
patient-years for epoetin beta, and 0.2 case per subsequent inhibition of erythropoietin-receptor
100,000 patient-years for the formulation of epoe- signaling resulted in inhibition of angiogenesis and
tin alfa in Epogen (Amgen). After procedures were destruction of tumor masses.87
adopted to ensure appropriate storage, handling, A recent study investigating the effect of therapy
and administration of Eprex to patients with chron- with epoetin on the clinical course of patients with
ic kidney disease, the exposure-adjusted incidence metastatic breast carcinoma was discontinued be-
decreased by 83 percent worldwide. cause of a trend toward higher mortality among pa-
Although the positive short-term effects of ther- tients receiving the drug.93 Controversy concerning
apy with erythropoietic agents on the correction of the use of epoetin in patients with cancer who have
anemia and avoidance of blood transfusions are well anemia of chronic disease has also arisen in two
documented,14,60,76 few data are available on pos- studies involving patients with head and neck tu-
sible effects on the course of underlying disease, mors. In one study, the increase in hemoglobin lev-
particularly since epoetin can exert additional bio- els with epoetin therapy was associated with a

n engl j med 352;10 www.nejm.org march 10, 2005 1019


The new england journal of medicine

Monitoring Therapy
Before the initiation of therapy with an erythropoi-
etic agent, iron deficiency should be ruled out (Fig.
2). For monitoring the response to erythropoietic
agents, hemoglobin levels should be determined af-
ter four weeks of therapy and at intervals of two to
four weeks thereafter. If the hemoglobin level in-
creases by less than 1 g per deciliter, the iron status
should be reevaluated (Table 2) and iron supple-
mentation considered.14,63 If iron-restricted eryth-
ropoiesis is not present, a 50 percent escalation in
the dose of the erythropoietic agent is indicated. The
dose of the erythropoietic agent should be adjusted
once the hemoglobin concentration reaches 12 g
per deciliter.14,62 If no response is achieved after
eight weeks of optimal dosage in the absence of iron
deficiency, a patient is considered nonresponsive
to erythropoietic agents.

conclusions
Advances in our understanding of the pathophysi-
ology of anemia of chronic disease including dis-
turbances of iron homeostasis, impaired prolifera-
tion of erythroid progenitor cells, and a blunted
erythropoietin response to anemia have made
possible the emergence of new therapeutic strate-
Figure 2. Algorithm for the Differential Diagnosis among Iron-Deficiency
gies. These include treatment of the underlying dis-
Anemia, Anemia of Chronic Disease, and Anemia of Chronic Disease ease and the use of erythropoietic agents, iron, or
with Iron Deficiency. blood transfusions. Needed are prospective, con-
The abbreviation sTfR/log ferritin denotes the ratio of the concentration of trolled studies to evaluate the effect of the manage-
soluble transferrin receptor to the log of the serum ferritin level in convention- ment of anemia on underlying diseases, with de-
al units. fined end points and analysis of the possible clinical
significance of erythropoietin-receptor expression
on certain tumor cells. Future strategies may include
favorable clinical outcome, improved tumor oxy- the use of iron-chelation therapy to induce the en-
genation, and increased susceptibility of tumors dogenous formation of erythropoietin,96 hepcidin
to preoperative chemoradiation therapy.94 In con- antagonists that overcome the retention of iron
trast, a double-blind prospective study investigat- within the reticuloendothelial system, and hor-
ing whether target hemoglobin levels greater than mones or cytokines that might effectively stimulate
13 g per deciliter for women and greater than 14 g erythropoiesis under inflammatory conditions. End
per deciliter for men improved regional tumor con- points that correlate with improvements in morbid-
trol among patients undergoing radiation therapy ity and mortality in well-designed, prospective stud-
for squamous-cell carcinoma of the head or neck ies must be identified in order to determine the op-
showed a recurrence rate among patients who were timal therapeutic regimen for patients with anemia
treated with epoetin that was higher than that of chronic disease.
among patients treated with placebo.95 Supported by grants from the Austrian Research Funds, FWF.
Current findings indicate that for patients receiv- Dr. Goodnough reports that he holds uncompensated positions
as vice president of the National Anemia Action Council and presi-
ing erythropoietic agents, target hemoglobin levels dent of the Society for the advancement of Blood Management;
should be 11 to 12 g per deciliter.14,62,63 Overcor- these organizations are supported by grants from a number of com-
rection of anemia to normal hemoglobin levels95 panies that produce products in this therapeutic area. He also re-
ports having received lecture fees from Amgen, Ortho Biotech, Wat-
and insufficient treatment64 have each been associ- son Pharmaceuticals, American Regent, and KV Pharmaceuticals.
ated with unfavorable clinical courses.

1020 n engl j med 352;10 www.nejm.org march 10, 2005


medical progress

references
1. Cartwright GE. The anemia of chronic ciated anemia. Am J Gastroenterol 2001;96: 30. Moura E, Noordermeer MA, Verhoeven
disorders. Semin Hematol 1966;3:351-75. 2382-7. N, Verheul AF, Marx JJ. Iron release from hu-
2. Matzner Y, Levy S, Grossowicz N, Izak 16. Wilson A, Reyes E, Ofman J. Prevalence man monocytes after erythrophagocytosis
G, Hershko C. Prevalence and causes of ane- and outcomes of anemia in inflammatory in vitro: an investigation in normal subjects
mia in elderly hospitalized patients. Geron- bowel disease: a systematic review of the lit- and hereditary hemochromatosis patients.
tology 1979;25:113-9. erature. Am J Med 2004;116:Suppl 7A:44S- Blood 1998;92:2511-9.
3. Weiss G. Pathogenesis and treatment of 49S. 31. Andrews NC. The iron transporter
anaemia of chronic disease. Blood Rev 17. Muller HM, Horina JH, Kniepeiss D, et DMT1. Int J Biochem Cell Biol 1999;31:991-
2002;16:87-96. al. Characteristics and clinical relevance of 4.
4. Means RT Jr. Recent developments in chronic anemia in adult heart transplant re- 32. Ludwiczek S, Aigner E, Theurl I, Weiss
the anemia of chronic disease. Curr Hema- cipients. Clin Transplant 2001;15:343-8. G. Cytokine-mediated regulation of iron
tol Rep 2003;2:116-21. 18. Frost AE, Keller CA. Anemia and eryth- transport in human monocytic cells. Blood
5. Maury CP, Liljestrom M, Laiho K, Tiitin- ropoietin levels in recipients of solid organ 2003;101:4148-54.
en S, Kaarela K, Hurme M. Tumor necrosis transplants. Transplantation 1993;56:1008- 33. Pietrangelo A. Physiology of iron trans-
factor alpha, its soluble receptor I, and -308 11. port and the hemochromatosis gene. Am J
gene promoter polymorphism in patients 19. Maheshwari A, Mishra R, Thuluvath PJ. Physiol Gastrointest Liver Physiol 2002;282:
with rheumatoid arthritis with or without Post-liver-transplant anemia: etiology and G403-G414.
amyloidosis: implications for the pathogen- management. Liver Transpl 2004;10:165- 34. Tilg H, Ulmer H, Kaser A, Weiss G. Role
esis of nephropathy and anemia of chronic 73. of IL-10 for induction of anemia during in-
disease in reactive amyloidosis. Arthritis 20. Collins AJ, Li S, St Peter W, et al. Death, flammation. J Immunol 2002;169:2204-9.
Rheum 2003;48:3068-76. hospitalization, and economic associations 35. Nemeth E, Rivera S, Gabayan V, et al.
6. Denz H, Huber P, Landmann R, Orth B, among incident hemodialysis patients with IL-6 mediates hypoferremia of inflamma-
Wachter H, Fuchs D. Association between hematocrit values of 36 to 39%. J Am Soc tion by inducing the synthesis of the iron
the activation of macrophages, changes of Nephrol 2001;12:2465-73. regulatory hormone hepcidin. J Clin Invest
iron metabolism and the degree of anaemia 21. Locatelli F, Pisoni RL, Combe C, et al. 2004;113:1271-6.
in patients with malignant disorders. Eur J Anaemia in haemodialysis patients of five 36. Nicolas G, Bennoun M, Porteu A, et al.
Haematol 1992;48:244-8. European countries: association with mor- Severe iron deficiency anemia in transgenic
7. Guralnik JM, Eisenstaedt RS, Ferrucci L, bidity and mortality in the Dialysis Out- mice expressing liver hepcidin. Proc Natl
Klein HG, Woodman RC. The prevalence of comes and Practice Patterns Study (DOPPS). Acad Sci U S A 2002;99:4596-601.
anemia in persons age 65 and older in the Nephrol Dial Transplant 2004;19:121-32. 37. Laftah AH, Ramesh B, Simpson RJ, et al.
United States: evidence for a high rate of un- [Erratum, Nephron Dial Transplant 2004; Effect of hepcidin on intestinal iron absorp-
explained anemia. Blood 2004;104:2263-8. 19:1666.] tion in mice. Blood 2004;103:3940-4.
8. Sullivan PS, Hanson DL, Chu SY, Jones 22. Stenvinkel P. The role of inflammation 38. Andrews NC. Anemia of inflammation:
JL, Ward JW. Epidemiology of anemia in hu- in the anaemia of end-stage renal disease. the cytokine-hepcidin link. J Clin Invest
man immunodeficiency virus (HIV)-infect- Nephrol Dial Transplant 2001;16:Suppl 7: 2004;113:1251-3.
ed persons: results from the multistate adult 36-40. 39. Papanikolaou G, Samuels ME, Ludwig
and adolescent spectrum of HIV disease sur- 23. Yap GS, Stevenson MM. Inhibition of in EH, et al. Mutations in HFE2 cause iron
veillance project. Blood 1998;91:301-8. vitro erythropoiesis by soluble mediators in overload in chromosome 1q-linked juvenile
9. Nissenson AR, Goodnough LT, Dubois Plasmodium chabaudi AS malaria: lack of a hemochromatosis. Nat Genet 2004;36:77-
RW. Anemia: not just an innocent bystand- major role for interleukin 1, tumor necrosis 82.
er? Arch Intern Med 2003;163:1400-4. [Er- factor alpha, and gamma interferon. Infect 40. Wang CQ, Udupa KB, Lipschitz DA. In-
ratum, Arch Intern Med 2003;163:1820.] Immun 1994;62:357-62. terferon-gamma exerts its negative regula-
10. van Iperen CE, van de Wiel A, Marx JJ. 24. Gordeuk VR, Delanghe JR, Langlois tory effect primarily on the earliest stages of
Acute event-related anaemia. Br J Haematol MR, Boelaert JR. Iron status and the out- murine erythroid progenitor cell develop-
2001;115:739-43. come of HIV infection: an overview. J Clin ment. J Cell Physiol 1995;162:134-8.
11. Dunn A, Carter J, Carter H. Anemia at Virol 2001;20:111-5. 41. Taniguchi S, Dai CH, Price JO, Krantz
the end of life: prevalence, significance, and 25. Rodriguez RM, Corwin HL, Gettinger SB. Interferon gamma downregulates stem
causes in patients receiving palliative care. A, Corwin MJ, Gubler D, Pearl RG. Nutri- cell factor and erythropoietin receptors but
J Pain Symptom Manage 2003;26:1132-9. tional deficiencies and blunted erythropoi- not insulin-like growth factor-I receptors in
12. Harrison L, Shasha D, Shiaova L, White etin response as causes of the anemia of crit- human erythroid colony-forming cells.
C, Ramdeen B, Portenoy R. Prevalence of ical illness. J Crit Care 2001;16:36-41. Blood 1997;90:2244-52.
anemia in cancer patients undergoing radia- 26. Groopman JE, Itri LM. Chemotherapy- 42. Maciejewski JP, Selleri C, Sato T, et al.
tion therapy. Semin Oncol 2001;28:54-9. induced anemia in adults: incidence and Nitric oxide suppression of human hemato-
13. Ludwig H, Fritz E, Leitgeb C, Pecher- treatment. J Natl Cancer Inst 1999;91:1616- poiesis in vitro: contribution to inhibitory
storfer M, Samonigg H, Schuster J. Predic- 34. [Erratum, J Natl Cancer Inst 2000;92: action of interferon-gamma and tumor ne-
tion of response to erythropoietin treatment 497.] crosis factor-alpha. J Clin Invest 1995;96:
in chronic anemia of cancer. Blood 1994;84: 27. Eschbach JW. Anemia management in 1085-92.
1056-63. chronic kidney disease: role of factors af- 43. Miller CB, Jones RJ, Piantadosi S, Abe-
14. Rizzo JD, Lichtin AE, Woolf SH, et al. fecting epoetin responsiveness. J Am Soc loff MD, Spivak JL. Decreased erythropoi-
Use of epoetin in patients with cancer: evi- Nephrol 2002;13:1412-4. etin response in patients with the anemia of
dence-based clinical practice guidelines of 28. Alvarez-Hernandez X, Liceaga J, McKay cancer. N Engl J Med 1990;322:1689-92.
the American Society of Clinical Oncology IC, Brock JH. Induction of hypoferremia and 44. Cazzola M, Ponchio L, de Benedetti F, et
and the American Society of Hematology. modulation of macrophage iron metabo- al. Defective iron supply for erythropoiesis
J Clin Oncol 2002;20:4083-107. lism by tumor necrosis factor. Lab Invest and adequate endogenous erythropoietin
15. Gasche C, Waldhoer T, Feichtenschlag- 1989;61:319-22. production in the anemia associated with
er T, et al. Prediction of response to iron su- 29. Torti FM, Torti SV. Regulation of ferritin systemic-onset juvenile chronic arthritis.
crose in inflammatory bowel disease-asso- genes and protein. Blood 2002;99:3505-16. Blood 1996;87:4824-30.

n engl j med 352;10 www.nejm.org march 10, 2005 1021


The new england journal of medicine

45. Jelkmann W. Proinflammatory cyto- num chemotherapy: results of a random- Intravenous iron optimizes the response to
kines lowering erythropoietin production. ized, double-blind, placebo-controlled trial. recombinant human erythropoietin in can-
J Interferon Cytokine Res 1998;18:555-9. J Clin Oncol 2001;19:2865-74. cer patients with chemotherapy-related ane-
46. Means RT Jr, Krantz SB. Inhibition of 61. Ma JZ, Ebben J, Xia H, Collins AJ. Hema- mia: a multicenter, open-label, randomized
human erythroid colony-forming units by tocrit level and associated mortality in he- trial. J Clin Oncol 2004;22:1301-7.
gamma interferon can be corrected by re- modialysis patients. J Am Soc Nephrol 78. Stevens RG, Jones DY, Micozzi MS, Tay-
combinant human erythropoietin. Blood 1999;10:610-9. lor PR. Body iron stores and the risk of can-
1991;78:2564-7. 62. NKF-K/DOQI Clinical Practice Guide- cer. N Engl J Med 1988;319:1047-52.
47. Minoo P, Zadeh MM, Rottapel R, Lebrun lines for anemia of chronic kidney disease: 79. Jiang R, Manson JE, Meigs JB, Ma J, Rifai
JJ, Ali S. A novel SHP-1/Grb2-dependent update 2000. Am J Kidney Dis 2001;37: N, Hu FB. Body iron stores in relation to risk
mechanism of negative regulation of cyto- Suppl 1:S182-S238. [Erratum, Am J Kidney of type 2 diabetes in apparently healthy
kine-receptor signaling: contribution of Dis 2001;38:442.] women. JAMA 2004;291:711-7.
SHP-1 C-terminal tyrosines in cytokine sig- 63. Winn RJ. The NCCN guidelines devel- 80. Thompson JA, Gilliland DG, Prchal JT,
naling. Blood 2004;103:1398-407. opment process and infrastructure. Oncolo- et al. Effect of recombinant human erythro-
48. Moldawer LL, Marano MA, Wei H, et al. gy (Huntingt) 2000;14:26-30. poietin combined with granulocyte/macro-
Cachectin/tumor necrosis factor-alpha al- 64. Besarab A, Bolton WK, Browne JK, et al. phage colony-stimulating factor in the treat-
ters red blood cell kinetics and induces ane- The effects of normal as compared with low ment of patients with myelodysplastic
mia in vivo. FASEB J 1989;3:1637-43. hematocrit values in patients with cardiac syndrome. Blood 2000;95:1175-9.
49. Spivak JL. Iron and the anemia of chron- disease who are receiving hemodialysis and 81. Ludwig H, Fritz E, Kotzmann H, Hocker
ic disease. Oncology (Huntingt) 2002;16: epoetin. N Engl J Med 1998;339:584-90. P, Gisslinger H, Barnas U. Erythropoietin
Suppl 10:25-33. 65. Kletzmayr J, Sunder-Plassmann G, Horl treatment of anemia associated with multi-
50. Lipschitz DA, Cook JD, Finch CA. A clin- WH. High dose intravenous iron: a note of ple myeloma. N Engl J Med 1990;322:1693-
ical evaluation of serum ferritin as an index caution. Nephrol Dial Transplant 2002;17: 9.
of iron stores. N Engl J Med 1974;290:1213- 962-5. 82. Cooper AC, Mikhail A, Lethbridge MW,
6. 66. Sullivan JL. Iron therapy and cardiovas- Kemeny DM, Macdougall IC. Increased ex-
51. Mast AE, Blinder MA, Gronowski AM, cular disease. Kidney Int Suppl 1999;69: pression of erythropoiesis inhibiting cyto-
Chumley C, Scott MG. Clinical utility of the S135-S137. kines (IFN-gamma, TNF-alpha, IL-10, and
soluble transferrin receptor and compari- 67. Adamson JW, Eschbach JW. Erythropoi- IL-13) by T cells in patients exhibiting a poor
son with serum ferritin in several popula- etin for end-stage renal disease. N Engl J response to erythropoietin therapy. J Am Soc
tions. Clin Chem 1998;44:45-51. Med 1998;339:625-7. Nephrol 2003;14:1776-84.
52. Punnonen K, Irjala K, Rajamaki A. Se- 68. Moreland LW, Baumgartner SW, Schiff 83. Cella D, Dobrez D, Glaspy J. Control of
rum transferrin receptor and its ratio to se- MH, et al. Treatment of rheumatoid arthritis cancer-related anemia with erythropoietic
rum ferritin in the diagnosis of iron defi- with a recombinant human tumor necrosis agents: a review of evidence for improved
ciency. Blood 1997;89:1052-7. factor receptor (p75)-Fc fusion protein. quality of life and clinical outcomes. Ann
53. Weiss G. Iron and immunity: a double- N Engl J Med 1997;337:141-7. Oncol 2003;14:511-9.
edged sword. Eur J Clin Invest 2002;32: 69. Goodnough LT, Bach RG. Anemia, 84. Bennett CL, Luminari S, Nissenson AR,
Suppl 1:70-8. transfusion, and mortality. N Engl J Med et al. Pure red-cell aplasia and epoetin thera-
54. Brugnara C. Iron deficiency and eryth- 2001;345:1272-4. py. N Engl J Med 2004;351:1403-8.
ropoiesis: new diagnostic approaches. Clin 70. Vincent JL, Baron JF, Reinhart K, et al. 85. Aguilera A, Bajo MA, Diez JJ, et al. Ef-
Chem 2003;49:1573-8. Anemia and blood transfusion in critically ill fects of human recombinant erythropoietin
55. Beguin Y, Clemons GK, Pootrakul P, Fil- patients. JAMA 2002;288:1499-507. on inflammatory status in peritoneal dialy-
let G. Quantitative assessment of erythro- 71. Vamvakas EC, Blajchman MA. Deleteri- sis patients. Adv Perit Dial 2002;18:200-5.
poiesis and functional classification of ane- ous clinical effects of transfusion-associat- 86. Arcasoy MO, Amin K, Karayal AF, et al.
mia based on measurements of serum ed immunomodulation: fact or fiction? Functional significance of erythropoietin re-
transferrin receptor and erythropoietin. Blood 2001;97:1180-95. ceptor expression in breast cancer. Lab In-
Blood 1993;81:1067-76. 72. Weinberg ED. Iron loading and disease vest 2002;82:911-8.
56. Barosi G. Inadequate erythropoietin re- surveillance. Emerg Infect Dis 1999;5:346- 87. Yasuda Y, Fujita Y, Matsuo T, et al. Eryth-
sponse to anemia: definition and clinical 52. ropoietin regulates tumour growth of hu-
relevance. Ann Hematol 1994;68:215-23. 73. Teehan GS, Bahdouch D, Ruthazer R, man malignancies. Carcinogenesis 2003;
57. Henry D, Abels R, Larholt K. Prediction Balakrishnan VS, Snydman DR, Jaber BL. 24:1021-9. [Erratum, Carcinogenesis 2003;
of response to recombinant human erythro- Iron storage indices: novel predictors of 24:1567.]
poietin (r-HuEPO/epoetin-alpha) therapy in bacteremia in hemodialysis patients initiat- 88. Acs G, Zhang PJ, McGrath CM, et al. Hy-
cancer patients. Blood 1995;85:1676-8. ing intravenous iron therapy. Clin Infect Dis poxia-inducible erythropoietin signaling in
58. Murphy ST, Parfrey PS. The impact of 2004;38:1090-4. squamous dysplasia and squamous cell car-
anemia correction on cardiovascular disease 74. Kaltwasser JP, Kessler U, Gottschalk R, cinoma of the uterine cervix and its potential
in end-stage renal disease. Semin Nephrol Stucki G, Moller B. Effect of recombinant role in cervical carcinogenesis and tumor
2000;20:350-5. human erythropoietin and intravenous iron progression. Am J Pathol 2003;162:1789-
59. Moreno F, Sanz-Guajardo D, Lopez- on anemia and disease activity in rheuma- 806.
Gomez JM, Jofre R, Valderrabano F. Increas- toid arthritis. J Rheumatol 2001;28:2430-6. 89. Mittelman M, Neumann D, Peled A,
ing the hematocrit has a beneficial effect on 75. Weiss G, Meusburger E, Radacher G, Kanter P, Haran-Ghera N. Erythropoietin in-
quality of life and is safe in selected hemodi- Garimorth K, Neyer U, Mayer G. Effect of duces tumor regression and antitumor im-
alysis patients: Spanish Cooperative Renal iron treatment on circulating cytokine levels mune responses in murine myeloma mod-
Patients Quality of Life Study Group of the in ESRD patients receiving recombinant hu- els. Proc Natl Acad Sci U S A 2001;98:5181-6.
Spanish Society of Nephrology. J Am Soc man erythropoietin. Kidney Int 2003;64: 90. Westenfelder C, Baranowski RL. Eryth-
Nephrol 2000;11:335-42. 572-8. ropoietin stimulates proliferation of human
60. Littlewood TJ, Bajetta E, Nortier JW, Ver- 76. Goodnough LT, Skikne B, Brugnara C. renal carcinoma cells. Kidney Int 2000;58:
cammen E, Rapoport B. Effects of epoetin Erythropoietin, iron, and erythropoiesis. 647-57.
alfa on hematologic parameters and quality Blood 2000;96:823-33. 91. Acs G, Acs P, Beckwith SM, et al. Eryth-
of life in cancer patients receiving nonplati- 77. Auerbach M, Ballard H, Trout JR, et al. ropoietin and erythropoietin receptor ex-

1022 n engl j med 352;10 www.nejm.org march 10, 2005


medical progress

pression in human cancer. Cancer Res 2001; 94. Glaser CM, Millesi W, Kornek GV, et al. apy: randomised, double-blind, placebo-
61:3561-5. Impact of hemoglobin level and use of re- controlled trial. Lancet 2003;362:1255-60.
92. Heeschen C, Aicher A, Lehmann R, et al. combinant erythropoietin on efficacy of pre- 96. Salvarani C, Baricchi R, Lasagni D, et al.
Erythropoietin is a potent physiologic stim- operative chemoradiation therapy for squa- Effects of desferrioxamine therapy on chron-
ulus for endothelial progenitor cell mobili- mous cell carcinoma of the oral cavity and ic disease anemia associated with rheuma-
zation. Blood 2003;102:1340-6. [Erratum, oropharynx. Int J Radiat Oncol Biol Phys toid arthritis. Rheumatol Int 1996;16:45-8.
Blood 2004;103:4388.] 2001;50:705-15. Copyright 2005 Massachusetts Medical Society.
93. Leyland-Jones B. Breast cancer trial with 95. Henke M, Laszig R, Rube C, et al.
erythropoietin terminated unexpectedly. Erythropoietin to treat head and neck cancer
Lancet Oncol 2003;4:459-60. patients with anaemia undergoing radiother-

n engl j med 352;10 www.nejm.org march 10, 2005 1023


Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

Das könnte Ihnen auch gefallen