Sie sind auf Seite 1von 10

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Dan L. Longo, M.D., Editor

Anemia of Inflammation
Tomas Ganz, Ph.D., M.D.​​

A
From the Departments of Medicine and nemia of inflammation (also referred to as anemia of chronic
Pathology, David Geffen School of Medi- disease) has been recognized for more than 60 years as a mild to moder-
cine at UCLA, Los Angeles. Address reprint
requests to Dr. Ganz at the David Geffen ately severe anemia (hemoglobin level, 7 to 12 g per deciliter) that develops
School of Medicine at UCLA, 43-229 CHS, in the context of systemic inflammation because of decreased production of erythro-
Box 951690, 10833 LeConte Ave., Los cytes, accompanied by a modest reduction in erythrocyte survival.1 The disorder,
­Angeles, CA 90095, or at ­tganz@​­mednet​
.­ucla​.­edu. like iron-deficiency anemia, is characterized by low serum iron levels (hypofer-
remia), but it differs from iron-deficiency anemia in that iron stores are preserved
N Engl J Med 2019;381:1148-57.
DOI: 10.1056/NEJMra1804281 in marrow macrophages, as well as in splenic and hepatic macrophages that re-
Copyright © 2019 Massachusetts Medical Society. cycle senescent erythrocytes. Thus, anemia of inflammation is primarily a disorder
of iron distribution.1
Whereas erythrocytes in iron-deficiency anemia are often small (low mean
corpuscular volume) and hemoglobin-deficient (low mean corpuscular hemoglobin
concentration), the erythrocytes in anemia of inflammation most often appear
normal, although they become small and pale in a subset of patients, particularly
those in whom iron deficiency coexists or develops as a complication. The normal
lifespan of human erythrocytes is about 120 days, and even if the lifespan is some-
what decreased by inflammation, a clinically significant decrement in the erythro-
cyte count does not usually develop until weeks to months after the onset of the
underlying inflammatory disorder, accounting for the association of anemia of in-
flammation with chronic diseases. In critically ill patients, however, anemia of
inflammation can develop very rapidly, after 1 week in a critical care unit.2 In such
patients, the progression of anemia of inflammation is often accelerated by con-
comitant blood loss (iatrogenic or disease-associated) or hemolysis, processes that
rapidly unmask the suppressive effect of inflammation on the erythropoietic sys-
tem and its inability to augment erythropoiesis in response to the loss of erythro-
cytes.3,4
Anemia of inflammation and iron-deficiency anemia are the two most common
anemias worldwide, and they are often coexisting disorders in people who live in
developing countries with a high prevalence of nutritional deficiencies and infec-
tions.5 In populations with better access to medical care and iron-rich nutrients,
anemia of inflammation is classically associated with chronic systemic inflamma-
tory disorders, including rheumatoid arthritis and systemic lupus erythematosus;
inflammatory bowel disease; chronic infections, including tuberculosis and the
acquired immunodeficiency syndrome; hematologic cancers associated with in-
creased cytokine production, such as Hodgkin’s lymphoma and some types of
non-Hodgkin’s lymphoma; and certain solid tumors (e.g., ovarian cancer and lung
cancer). Increasingly, anemia of inflammation is recognized as the main or con-
tributing cause of anemia in many other patients with systemic inflammation,
including those with chronic kidney disease,6 chronic heart failure,7 chronic ob-
structive pulmonary disease,8 or cystic fibrosis.9

1148 n engl j med 381;12 nejm.org  September 19, 2019

The New England Journal of Medicine


Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Anemia of Inflammation

Infl a m m at ion storing hepatocytes. Plasma iron is bound to the


transport protein transferrin and is mostly des-
Inflammation is a set of biologic mechanisms tined for delivery to marrow erythroblasts that
that evolved in multicellular organisms to con- consume iron to synthesize heme and hemoglo-
strain and neutralize infectious and other injurious bin, with smaller iron flows meeting the iron
agents that entered the tissues. The initial recog- requirements of all other organs and tissues.
nition of such agents is mediated by molecular Baseline hepcidin synthesis is controlled by feed-
sensors on membranes of host defense cells, in back from both iron stores and plasma iron levels.
cellular cytoplasm, or in extracellular fluid (e.g., The hepatic iron regulatory system is based
toll-like receptors, Nod-like receptors, and man- on complex and incompletely understood inter-
nose-binding lectin). On contact with specific actions between at least two types of iron sen-
molecular patterns that are characteristic of in- sors in the liver that interact with the hepatocyte
fectious or injurious agents, the activation of bone morphogenetic protein receptor and affect
these molecular sensors generates mediators that the signaling pathway that transcriptionally regu-
eventually give rise to the clinically detectable lates hepcidin synthesis.12 The two types of iron
manifestations of inflammation. In addition, in- sensors detect the level of diferric transferrin in
flammation may develop as a result of immune plasma (transferrin receptors 1 and 2) and the
dysregulation in autoimmune or malignant dis- amount of iron stored in the liver (sensor not yet
orders (e.g., rheumatologic disorders, inflamma- identified), respectively. These iron-sensing mech-
tory bowel disease, and Hodgkin’s lymphoma). anisms increase hepcidin synthesis when iron
Whether the aging process itself causes dysregu- levels are abundant and decrease it during iron
lation of inflammation, even in the absence of a deficiency, thereby modulating dietary iron ab-
specific inflammatory disease, remains contro- sorption and release from stores in response to
versial.10 Cytokines, including tumor necrosis fac- iron status. Inflammation greatly increases hep-
tor α (TNF-α), interleukin-1, interleukin-6, and cidin synthesis, which is important in the patho-
interferon-γ, are produced by inflammatory cells genesis of anemia of inflammation. Hepcidin
within the first hours after the onset of inflam- synthesis increases predominantly but not exclu-
mation; these cytokines restrict erythropoiesis sively13 because of interleukin-6,14 which acts
both directly and indirectly and shorten the through the JAK2–STAT3 (Janus kinase 2–signal
erythrocyte lifespan. transducer and activator of transcription 3) path-
way to increase the transcription of the hepcidin
gene in hepatocytes. Although small but clini-
Iron Me ta bol ism
cally relevant increases in serum hepcidin develop
During normal erythropoiesis, when there is an even in relatively mild systemic inflammatory
adequate nutritional iron supply and an absence states, such as obesity,15 serum hepcidin levels
of inflammation, systemic iron homeostasis may increase by a factor of more than 10 in
maintains plasma iron levels in the range of 10 patients with sepsis as compared with healthy
to 30 μM and whole-body iron stores in the range persons.16
of 0.3 to 1 g, with the lower end of the ranges
typical for women of reproductive age. The main Er y throp oie sis
mechanism of iron homeostasis centers on the
interaction between the iron regulatory hormone Erythrocytes are generated from hematopoietic
hepcidin, produced by hepatocytes, and ferro- stem cells in the marrow through steps involving
portin, which is both the hepcidin receptor and progressively more restricted lineage choices17
the sole cellular iron exporter through which iron and decreasing proliferative capacity.18 Although
is transferred to blood plasma11 (Fig. 1). Hepci- the specific pathways controlling these lineage
din inhibits the iron-exporting activity of ferro- choices are still not fully understood,19 it appears
portin, thereby controlling the transfer of iron to that some populations of early progenitors have
blood plasma from iron-absorbing duodenal the potential for both myeloid differentiation and
enterocytes, from macrophages that recycle the erythroid or megakaryocytic differentiation, with
iron of senescent erythrocytes, and from iron- the path choice biased by mutually antagonistic

n engl j med 381;12 nejm.org  September 19, 2019 1149


The New England Journal of Medicine
Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Interleukin-6

Liver

Spleen
Hepcidin
Hepcidin _
Fpn
_ Fpn

↓Fe Iron-recycling
↓Fe macrophage
↓Red cells

Hepcidin ↓Plasma
Fe–Tf

Bone
Hepcidin marrow
Fpn clearance
_
↓Fe
Duodenum Impaired
renal
function

Figure 1. Changes in Iron Homeostasis in Anemia of Inflammation.


Iron restriction in anemia of inflammation is mediated by the interaction of increased levels of hepcidin with the
iron exporter ferroportin (Fpn). Hepcidin excess is exacerbated if renal function is impaired. Fe–Tf denotes iron–
transferrin complex.

interactions between two transcription factors: locytes, which are marked by their residual RNA
PU.1, which favors myeloid differentiation, and content. In blood, reticulocytes mature into bi-
GATA1, which favors erythroid differentiation.17 concave erythrocytes. The survival rate of CFU-Es
The relative proportions of the two transcription and early erythroblasts is determined by erythro-
factors can be influenced by inflammation. The poietin, and the differentiation and division of
first precursor fully committed to erythroid erythroblasts are controlled by the levels of eryth-
differentiation is BFU-E (burst-forming unit, ropoietin and iron,20 as well as by one or more
erythroid), defined by the ability of these cells activin-like members of the TGF-β (transform-
to proliferate and give rise to large clusters of ing growth factor β) superfamily.21 Erythropoiesis
erythroid cells when cultured in semisolid me- is dependent on the level of diferric transferrin
dium with appropriate growth factors. A more in plasma, a factor that is relevant to the patho-
differentiated cell type is CFU-E (colony-forming genesis of iron-restrictive anemias.
unit, erythroid), which generates fewer erythroid
cells under these conditions.18 CFU-Es give rise Patho gene sis
to even more differentiated proerythroblasts.
Each proerythroblast then undergoes four or five Evolutionary Perspective
more divisions, each of which yields progres- Responding to infection or injury by favoring
sively more mature hemoglobin-synthesizing host defense processes over housekeeping pro-
erythroblasts, finally enucleating to yield reticu- cesses such as erythropoiesis is a beneficial

1150 n engl j med 381;12 nejm.org September 19, 2019

The New England Journal of Medicine


Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Anemia of Inflammation

Inflammatory
stimulus
↑Myeloid
cells
Interferon-γ
Common
Interleukin-6
progenitor
TNF-α Interferon-γ
Hepcidin
↓Erythroid Hypoferremia ↓Iron release
precursors

↑Erythrophagocytosis
↓Proliferation
↓Erythrocyte lifespan

Bone marrow Blood and iron-recycling macrophages

Figure 2. Role of Systemic Inflammation in Anemia.


Systemic inflammation is characterized by high levels of cytokines that bias hematopoiesis toward myeloid-cell pro-
duction (large green arrow) rather than erythropoiesis (red arrow) (interferon-γ), inhibit erythroid-precursor prolif-
eration (tumor necrosis factor α [TNF-α]), activate macrophages for erythrophagocytosis and thereby shorten the
erythrocyte lifespan (interferon-γ), and inhibit the release of recycled iron from macrophages (interleukin-6 through
hepcidin), causing hypoferremia. Hypoferremia inhibits erythroblast proliferation. The dashed lines represent solu-
ble mediators that regulate erythropoiesis during inflammation.

evolutionary strategy that provides a useful frame- invaded during infections with extracellular bac-
work for understanding anemia of inflamma- teria, contain only 2 to 3 mg of iron, as com-
tion. Hypoferremia and increased production and pared with 3 to 4 g of total body iron, and their
activation of leukocytes serve host defense at the iron content turns over every few hours, making
expense of erythrocyte production and erythro- the plasma iron level potentially unstable when
cyte survival. Increased production of leukocytes the iron supply or demand changes. Erythropoi-
and hypoferremia diminish the number of ery- esis represents the main consumer of iron from
throid precursors, and macrophage activation plasma, and the recycling of senescent or dam-
shortens the erythrocyte lifespan (Fig. 2). The aged erythrocytes by macrophages is the main
long lifespan of erythrocytes buffers the conse- source of iron for plasma. Both are strongly af-
quences of decreased erythropoiesis during most fected by various inflammatory processes, mak-
acute infections. In chronic infections or inflam- ing tight control of the iron level during inflam-
matory disorders, erythrocyte counts are often mation particularly important for host defense.
reduced to an anemic steady state in which the High levels of circulating hepcidin, stimulated by
destruction of erythrocytes matches their de- interleukin-6, inhibit the export of cellular iron
creased production, a condition that has been into plasma, thereby decreasing both intestinal
very difficult to model in laboratory rodents. absorption of iron and the release of iron from
erythrocyte-recycling macrophages in the spleen
Hypoferremia and the liver. Macrophages and hepatocytes, the
Hypoferremia develops within the first few hours two cell types that accumulate iron, secrete large
after infection or other inflammatory events. amounts of ferritin through nonclassical secre-
The decreases in the plasma iron level and trans- tory mechanisms.24 Hypoferremia associated with
ferrin saturation may serve to prevent the genera- elevated plasma ferritin and hepcidin levels is
tion of nontransferrin-bound iron, an iron spe- characteristic of anemia of inflammation, where-
cies now recognized as a potent stimulus to the as in iron-deficiency anemia, hypoferremia asso-
pathogenicity of gram-negative bacteria22,23 and ciated with low plasma ferritin and hepcidin
probably other microbes as well. Plasma and other levels is characteristic.
extracellular fluid, which are the compartments Inflammatory hypoferremia, like the hypofer-

n engl j med 381;12 nejm.org September 19, 2019 1151


The New England Journal of Medicine
Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

remia of systemic iron deficiency, has an inhibi- rocyte lifespan, to approximately 90 days, has
tory effect on erythropoiesis. Iron is not simply been consistently shown, originally with the use
stoichiometrically limiting for hemoglobin syn- of radioactive iron tracers36 and currently by
thesis, in which each hemoglobin molecule re- means of measurements of exhaled carbon mon-
quires four iron atoms. Rather, erythropoiesis is oxide,37 a product of heme degradation. A de-
inhibited by hypoferremia25,26 at a relatively high creased erythrocyte lifespan is also seen in many
set point (transferrin saturation of 15 to 20%) in patients with inflammation who do not have
a manner that appears to protect the iron supply anemia,37 so anemia will develop only when the
for other tissues (e.g., muscle, central nervous erythropoietic compensation is impaired. The
system, and nonerythroid marrow), which are heterogeneity of underlying diseases in anemia
less affected by the decreased plasma iron level. of inflammation makes it likely that multiple
factors contribute to the increased destruction of
Bone Marrow Reprogramming erythrocytes, including macrophage activation28
Leukocytosis and increased production of leuko- (which may affect the threshold for recognition
cytes in the marrow are early inflammatory re- of erythrocyte senescence) and exposure of eryth-
sponses, manifested in the marrow by an increased rocytes to bystander inflammatory damage.38
number of myeloid precursors (ratio of myeloid The term “consumptive anemia of inflammation”
to erythroid precursors, >4:1). Such reprogram- refers to disorders in which hemophagocytosis
ming of the marrow is mediated by inflamma- by activated macrophages is the predominant
tory cytokines (e.g., TNF-α27 and interferon-γ,28 cause of anemia.39 Extrapolating from frank hemo-
which have been studied most extensively) that lytic anemias associated with infections and severe
activate the transcription factor PU.1 to promote systemic inflammation, erythrocytes are likely
myelopoiesis and lymphopoiesis at the expense to be damaged even in anemia of inflammation
of erythropoiesis. The capacity of BFU-E to give by the deposition of antibody and complement
rise to more differentiated erythroid cells is also and injury from microvascular fibrin strands.
inhibited by inflammatory cytokines.29,30
Another form of marrow reprogramming is Di agnosis of A nemi a
mediated by inflammatory suppression of eryth- of Infl a m m at ion
ropoietin, the master erythropoietic hormone.
Serum erythropoietin levels are reduced in a Symptoms of mild-to-moderate anemia in pa-
subgroup of patients with systemic inflamma- tients with anemia of inflammation include
tion,31,32 at least as compared with patients whofatigue, exercise intolerance, and exertional dys-
have a similar degree of iron-deficiency anemia. pnea, but these symptoms are difficult to distin-
The plausibility of a mechanism that is dependentguish from the effects of chronic systemic inflam-
on the effect of inflammation on erythropoietin- mation. Anemia of inflammation is diagnosed
producing renal interstitial cells is supported by
in patients with normocytic and normochromic
experiments in murine models.33 The responsive- anemia (normal mean corpuscular volume and
ness of erythroid precursors to erythropoietin isnormal mean corpuscular hemoglobin concen-
also impaired by inflammation, as commonly tration, respectively) in whom there is evidence
manifested by increased exogenous erythropoie- of systemic inflammation (increased erythrocyte
tin requirements in patients with end-stage kid- sedimentation rate or C-reactive protein level) and
ney disease who have inflammation.34,35 The re- evidence of iron restriction that is not caused by
sistance to erythropoietin is mediated in part systemic iron deficiency (low transferrin satura-
through a decrease in the number of erythropoi- tion [<20%] along with a high serum ferritin
etin receptors on erythroid progenitors, which islevel [>100 μg per liter]). The main challenge in
a recently described effect of hypoferremia that establishing a specific diagnosis is the common
results in a diminished proliferative capacity ofcoexistence of true iron deficiency and anemia of
these progenitors.20 inflammation (especially in patients with blood
loss from underlying disease) or an iron deficit
Shortened Erythrocyte Lifespan caused by malnutrition, long-standing inflam-
In patients with anemia of inflammation, a mod- mation, or increased iron requirements (in grow-
erate (approximately 25%) decrease in the eryth- ing children or pregnant women).

1152 n engl j med 381;12 nejm.org  September 19, 2019

The New England Journal of Medicine


Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Anemia of Inflammation

Differences in biomarkers of anemia of in- Table 1. Differences in Biomarkers of Iron Deficiency and Anemia
flammation and true iron deficiency are listed in of Inflammation.
Table 1. In practice, these markers are least help-
ful when they are most needed (i.e., when ane- Iron Anemia of
Biomarker* Deficiency Inflammation
mia of inflammation is complicated by coexist-
ing iron deficiency).40 The main challenges to Mean corpuscular volume Low Normal
developing widely applicable algorithms that Mean corpuscular hemoglobin Low Normal
would assess the contribution of iron deficiency Reticulocyte hemoglobin content Low Normal
in anemia of inflammation include the hetero- Percentage of hypochromic erythrocytes High Low
geneity of the underlying diseases, the variable Serum transferrin High Low
relative severity of the components of inflamma-
Serum transferrin receptor High Normal
tion and iron deficiency, and the lack of a stan-
dard test for evaluating the performance of these Serum ferritin Low High
markers individually or in combination. Histori- Serum hepcidin Low High
cally, the absence of stainable iron in the bone
* Intermediate biomarker values would be expected when both iron deficiency
marrow was used as a standard for the diagno- and anemia of inflammation are present.
sis of iron deficiency, but this test is inappropri-
ately invasive in most situations and is subject to
sampling problems and interpretation by pa- correct the anemia if it were caused solely by
thologists.41-43 It is therefore not useful in the iron deficiency and administer half this amount.
large clinical studies that would be required for A useful and easy-to-remember formula for this
the development of a generally applicable diag- estimate is as follows: iron (in milligrams) = hemo-
nostic algorithm for detecting a clinically rele- globin deficit (in grams per deciliter) × body weight
vant contribution of iron deficiency in patients (in pounds [convert kilograms to pounds by
with anemia of inflammation. multiplying by 2.2]). Partial hemoglobin correction
A practical clinical solution to the challenge typically occurs within 4 weeks after treatment
of diagnosing iron deficiency in patients with with intravenous iron and levels off by 8 weeks,
anemia of inflammation is to focus on the de- but patients may notice subjective improvement
tection and specific diagnosis of any occult blood much earlier. For one-time treatment, all currently
loss (most frequently gastrointestinal) and on any available intravenous iron preparations are simi-
iatrogenic blood loss from procedures and blood larly safe and effective, so the specific prepara-
sampling. A therapeutic trial of oral or intrave- tion and schedule of administration should be
nous iron can then be considered, depending on chosen on the basis of convenience for the pa-
the urgency of iron replacement and any side tient and total cost.46
effects of previous iron therapy, with the likely With respect to the potential effects of iron
effects of iron on both the underlying disease replacement on the underlying disease process,
process and the patient’s overall well-being taken active infection, especially with gram-negative
into account. bacteria, is of particular concern because of evi-
The absorption of oral iron is commonly im- dence from an animal model22 and clinical data47
paired in diseases with systemic inflammation,44 that implicate iron excess in the form of non-
although during mild inflammation, this effect transferrin-bound iron as an enhancer of micro-
is counteracted by the iron absorption–promoting bial pathogenicity. Indiscriminate iron supple-
effect of iron deficiency.45 In anemia of inflam- mentation may increase morbidity and mortality
mation, oral iron administration is therefore less in low-resource settings where infections are
reliable than the parenteral route and results in endemic and nutritional deficiencies are wide-
slower correction of the iron deficit. By contrast, spread.48
intravenous iron preparations have become safer
to administer and can be very effective in anemia T r e atmen t
of inflammation mixed with iron deficiency.32
One approach to intravenous iron therapy in pa- Treatments that target the infectious or inflam-
tients with both disorders is to estimate the matory processes that cause anemia of inflamma-
amount of iron that would be needed to fully tion will not only ameliorate the anemia but also

n engl j med 381;12 nejm.org  September 19, 2019 1153


The New England Journal of Medicine
Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

improve many symptoms and deficits caused by mild anemia in such patients but may be war-
the primary disease. Such treatments are there- ranted for more severe anemia (hemoglobin level,
fore more likely than treatments aimed solely at <10 g per deciliter). The use of these treatment
anemia of inflammation to yield a meaningful approaches in patients with anemia of inflam-
improvement for the patient. With such treat- mation who do not have chronic kidney disease
ments, anemia can be mitigated remarkably has been studied only in a very limited manner,56
rapidly, as documented in patients with Castle- and the disease-specific risks and benefits of
man’s disease after treatment with anti–interleu- erythropoietin therapy remain uncertain.
kin-6 antibody,49 patients with giant-cell arteritis In view of these limitations, the treatment of
after treatment with glucocorticoids,50 and pa- anemia of inflammation with erythropoietin,
tients with rheumatoid arthritis who received with or without intravenous iron, in patients who
anti–interleukin-6 or anti–TNF-α therapy,51 with do not have chronic kidney disease should be
clinically significant increases in hemoglobin considered on an individual basis, with the bene-
seen after 2 weeks of treatment. In patients with fits that would be meaningful to the patient
anemic tuberculosis, use of antimicrobial agents carefully weighed against the documented risks
completely resolved the anemia in about a third of these interventions, as defined in the studies
of the patients after 1 month of treatment and involving patients with chronic kidney disease
in about half the patients after 2 months of not requiring dialysis. As an example, consider
treatment.52 two patients, both with a hemoglobin level of
Unfortunately, effective treatment of the un- 8.5 g per deciliter. The patient who is hospital-
derlying inflammation is not always feasible. As ized with multiorgan manifestations of systemic
specific treatment options for anemia of inflam- vasculitis and urinary catheter–associated infec-
mation, erythropoietin derivatives, with or with- tion is easier to treat for anemia but much less
out intravenous iron, have been developed and likely to benefit meaningfully from the correc-
tested predominantly in patients with chronic tion of anemia than the patient who is a working
kidney disease. In addition to the effects of in- mother of two children and is receiving partially
flammation, such patients often have impairment effective therapy for inflammatory bowel disease.
in the expected anemia-stimulated increase in New compounds for the specific treatment of
renal production of erythropoietin. Although anemia of inflammation are under development.
there have been few specific clinical studies of The targeted mechanisms are designed to re-
the mechanism of action of erythropoietin de- verse hypoferremia in anemia of inflammation
rivatives combined with intravenous iron,53 the by decreasing the level of hepcidin with the use
combined drugs are expected to counteract the of hepcidin binders or by hindering the access of
iron-restrictive effect of inflammation, as well hepcidin to its target ferroportin or antagoniz-
as the associated resistance to endogenous ing the signaling pathways that stimulate hepci-
erythropoietin. din production during inflammation (Table 2).
A systematic review of the treatment of ane- Prolyl hydroxylase inhibitors stimulate the produc-
mia with erythropoietin derivatives in patients tion of erythropoietin but may also act directly on
with chronic kidney disease who did not require the intestinal mucosa to increase iron absorption
renal-replacement therapy showed a marked im- and thereby ameliorate iron restriction.
provement in blood hemoglobin levels and some
improvement in anemia-related symptoms. How- C on t r ibu t ion of A nemi a
ever, there was no measurable effect on the risk of Infl a m m at ion t o A dv er se
of disease progression requiring renal-replace- Ou t c ome s
ment therapy.54 The risks (chiefly treatment-­
related stroke) and some potential benefits of In Lifelines, a very large, prospective cohort
treatment with darbopoetin for patients with study, anemia of inflammation in older patients
chronic kidney disease and relatively mild ane- (>60 years of age) was associated with decreased
mia (similar in severity to anemia of inflamma- survival and impaired health-related quality of
tion) were recently analyzed and carefully con- life, most prominently, impaired physical func-
sidered.55 The authors concluded that treatment tioning.70 However, such studies cannot deter-
with darbopoetin is probably not advisable for mine whether anemia of inflammation is merely

1154 n engl j med 381;12 nejm.org  September 19, 2019

The New England Journal of Medicine


Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Anemia of Inflammation

Table 2. Compounds under Development for the Treatment of Anemia of Inflammation.

Compound Target* Stage of Development (Study)


Heparin derivatives BMPs that stimulate hepcidin synthesis Preclinical stage (Poli et al.57)
Soluble hemojuvelin BMPs that stimulate hepcidin synthesis Preclinical stage (Theurl et al.58)
Engineered hepcidin binders Hepcidin Preclinical stage (Hohlbaum et al.59) and
phase 1 (Boyce et al.60)
Monoclonal antibodies Ferroportin, blocking hepcidin access Phase 1 (Sheetz et al.61)
BMP-6 Phase 1 (Sheetz et al.61)
Hepcidin Phase 1 (Vadhan-Raj et al.62) and preclinical
stage (Sasu et al.63)
Hemojuvelin Preclinical stage (Kovac et al.64)
Momelotinib BMP receptor (ACVR1) and JAK1 Preclinical stage (Asshoff et al.65) and phase 3
and JAK2 (Mesa et al.66)†
TP-0184 BMP receptor (ACVR1) Phase 1 (Peterson et al.67)
Prolyl hydroxylase inhibitors HIF prolyl hydroxylases Preclinical stage (Barrett et al.68) and phase 2
(Chen et al.69)‡

* ACVR1 denotes activin A receptor 1, BMP bone morphogenetic protein, HIF hypoxia-inducible factor, and JAK1 and JAK2
Janus kinase 1 and 2, respectively.
† The phase 3 study involved patients with myelofibrosis.
‡ The phase 2 study involved patients with chronic kidney disease.

a marker of adverse outcomes or significantly improved outcomes that are meaningful to pa-
contributes to them.71 In principle, the contribu- tients. An increased understanding of the patho-
tion of anemia of inflammation to adverse out- genesis of anemia of inflammation has stimu-
comes can be ascertained only when highly lated the ongoing development of targeted
specific and safe treatments for the disorder therapies that may offer additional treatment
become available for clinical trials. options in the future.
Dr. Ganz reports receiving grant support, honoraria, and
Sum m a r y consulting fees from Keryx Biopharmaceuticals and Akebia
Therapeutics, being employed by and receiving nonfinancial
Anemia of inflammation is a highly prevalent support from Intrinsic LifeSciences, receiving consulting fees
from Vifor Fresenius, La Jolla Pharmaceutical, Sierra Oncology,
syndrome associated with systemic inflamma- and Ambys Medicines, receiving honoraria from Gilead Sci-
tion. In its most common form, anemia of in- ences, holding a pending patent application on erythroferrone
flammation is readily diagnosed as a normo- and Erfe polypeptides and methods of regulating iron metabolism
(US 2016/0122409 A1), assigned to Silarus Therapeutics, and
cytic, normochromic anemia associated with low holding a patent on competitive regulation of hepcidin mRNA
transferrin saturation but a high serum ferritin by soluble and cell-associated hemojuvelin (US 7534764B2). No
level. With currently available treatment ap- other potential conflict of interest relevant to this article was
reported.
proaches, therapeutic measures directed at the Disclosure forms provided by the author are available with the
underlying disease are most likely to result in full text of this article at NEJM.org.

References
1. Cartwright GE. The anemia of chron- 4. Corwin HL, Krantz SB. Anemia of the and hepcidin knockout mice. Am J Physiol
ic disorders. Semin Hematol 1966;​3:​351- critically ill: “acute” anemia of chronic Renal Physiol 2016;​311:​F1369-F1377.
75. disease. Crit Care Med 2000;​28:​3098-9. 7. Opasich C, Cazzola M, Scelsi L, et al.
2. Thomas J, Jensen L, Nahirniak S, Gib- 5. Shaw JG, Friedman JF. Iron deficiency Blunted erythropoietin production and
ney RTN. Anemia and blood transfusion anemia: focus on infectious diseases in defective iron supply for erythropoiesis as
practices in the critically ill: a prospective lesser developed countries. Anemia 2011;​ major causes of anaemia in patients with
cohort review. Heart Lung 2010;​ 39:​
217- 2011:​260380. chronic heart failure. Eur Heart J 2005;​26:​
25. 6. Hanudel MR, Chua K, Rappaport M, 2232-7.
3. Scharte M, Fink MP. Red blood cell et al. Effects of dietary iron intake and 8. Boutou AK, Hopkinson NS, Polkey
physiology in critical illness. Crit Care chronic kidney disease on fibroblast MI. Anaemia in chronic obstructive pul-
Med 2003;​31:​Suppl:​S651-S657. growth factor 23 metabolism in wild-type monary disease: an insight into its preva-

n engl j med 381;12 nejm.org  September 19, 2019 1155


The New England Journal of Medicine
Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

lence and pathophysiology. Clin Sci (Lond) Spektor L, et al. Ferritin is secreted via 38. Kim A, Fung E, Parikh SG, et al. A
2015;​128:​283-95. 2 distinct nonclassical vesicular pathways. mouse model of anemia of inflammation:
9. Fischer R, Simmerlein R, Huber RM, Blood 2018;​131:​342-52. complex pathogenesis with partial depen-
Schiffl H, Lang SM. Lung disease severity, 25. Bullock GC, Delehanty LL, Talbot AL, dence on hepcidin. Blood 2014;​123:​1129-
chronic inflammation, iron deficiency, and et al. Iron control of erythroid develop- 36.
erythropoietin response in adults with ment by a novel aconitase-associated 39. Zoller EE, Lykens JE, Terrell CE, et al.
cystic fibrosis. Pediatr Pulmonol 2007;​42:​ regulatory pathway. Blood 2010;​116:​97- Hemophagocytosis causes a consumptive
1193-7. 108. anemia of inflammation. J Exp Med 2011;​
10. Stauder R, Valent P, Theurl I. Anemia 26. Han AP, Yu C, Lu L, et al. Heme-regu- 208:​1203-14.
at older age: etiologies, clinical implica- lated eIF2alpha kinase (HRI) is required 40. Petzer V, Theurl I, Weiss G. Estab-
tions, and management. Blood 2018;​131:​ for translational regulation and survival lished and emerging concepts to treat im-
505-14. of erythroid precursors in iron deficiency. balances of iron homeostasis in inflam-
11. Ganz T. Systemic iron homeostasis. EMBO J 2001;​20:​6909-18. matory diseases. Pharmaceuticals (Basel)
Physiol Rev 2013;​93:​1721-41. 27. Orsini M, Chateauvieux S, Rhim J, et al. 2018;​11:​135.
12. Wang CY, Babitt JL. Liver iron sensing Sphingolipid-mediated inflammatory sig- 41. Thomason RW, Lavelle J, Nelson D,
and body iron homeostasis. Blood 2019;​ naling leading to autophagy inhibition Lin K, Uherova P. Parenteral iron therapy
133:​18-29. converts erythropoiesis to myelopoiesis is associated with a characteristic pattern
13. Roth MP, Meynard D, Coppin H. Regu- in human hematopoietic stem/progenitor of iron staining on bone marrow aspirate
lators of hepcidin expression. Vitam Horm cells. Cell Death Differ 2019;​26:​1796-812. smears. Am J Clin Pathol 2007;​128:​590-3.
2019;​110:​101-29. 28. Libregts SF, Gutiérrez L, de Bruin AM, 42. Barron BA, Hoyer JD, Tefferi A. A bone
14. Nemeth E, Rivera S, Gabayan V, et al. et al. Chronic IFN-γ production in mice marrow report of absent stainable iron is
IL-6 mediates hypoferremia of inflamma- induces anemia by reducing erythrocyte not diagnostic of iron deficiency. Ann
tion by inducing the synthesis of the iron life span and inhibiting erythropoiesis Hematol 2001;​80:​166-9.
regulatory hormone hepcidin. J Clin Invest through an IRF-1/PU.1 axis. Blood 2011;​ 43. Krause JR, Brubaker D, Kaplan S.
2004;​113:​1271-6. 118:​2578-88. Comparison of stainable iron in aspirated
15. Tussing-Humphreys LM, Nemeth E, 29. Means RT Jr, Dessypris EN, Krantz and needle-biopsy specimens of bone
Fantuzzi G, et al. Elevated systemic hepci- SB. Inhibition of human erythroid colony- marrow. Am J Clin Pathol 1979;​72:​68-70.
din and iron depletion in obese premeno- forming units by interleukin-1 is mediated 44. Weber J, Werre JM, Julius HW, Marx
pausal females. Obesity (Silver Spring) by gamma interferon. J Cell Physiol 1992;​ JJ. Decreased iron absorption in patients
2010;​18:​1449-56. 150:​59-64. with active rheumatoid arthritis, with and
16. Li H, Rose MJ, Tran L, et al. Develop- 30. Means RT Jr, Krantz SB. Inhibition of without iron deficiency. Ann Rheum Dis
ment of a method for the sensitive and human erythroid colony-forming units by 1988;​47:​404-9.
quantitative determination of hepcidin in gamma interferon can be corrected by re- 45. Stoffel NU, Lazrak M, Bellitir S, et al.
human serum using LC-MS/MS. J Phar- combinant human erythropoietin. Blood The opposing effects of acute inflamma-
macol Toxicol Methods 2009;​59:​171-80. 1991;​78:​2564-7. tion and iron deficiency anemia on serum
17. Orkin SH. Diversification of haemato- 31. Miller CB, Jones RJ, Piantadosi S, hepcidin and iron absorption in young
poietic stem cells to specific lineages. Nat Abeloff MD, Spivak JL. Decreased eryth- women. Haematologica 2019;​104:​1143-9.
Rev Genet 2000;​1:​57-64. ropoietin response in patients with the 46. Auerbach M, Macdougall I. The avail-
18. Dzierzak E, Philipsen S. Erythropoie- anemia of cancer. N Engl J Med 1990;​322:​ able intravenous iron formulations: his-
sis: development and differentiation. Cold 1689-92. tory, efficacy, and toxicology. Hemodial
Spring Harb Perspect Med 2013;​ 3(4):​ 32. Cazzola M, Ponchio L, de Benedetti F, Int 2017;​21:​Suppl 1:​S83-S92.
a011601. et al. Defective iron supply for erythropoi- 47. Belotti A, Duca L, Borin L, et al. Non
19. Dulmovits BM, Hom J, Narla A, Mo- esis and adequate endogenous erythro- transferrin bound iron (NTBI) in acute
handas N, Blanc L. Characterization, regu- poietin production in the anemia associ- leukemias throughout conventional inten-
lation, and targeting of erythroid progen- ated with systemic-onset juvenile chronic sive chemotherapy: kinetics of its appear-
itors in normal and disordered human arthritis. Blood 1996;​87:​4824-30. ance and potential predictive role in infec-
erythropoiesis. Curr Opin Hematol 2017;​ 33. Souma T, Yamazaki S, Moriguchi T, tious complications. Leuk Res 2015;​39:​
24:​159-66. et al. Plasticity of renal erythropoietin- 88-91.
20. Khalil S, Delehanty L, Grado S, et al. producing cells governs fibrosis. J Am Soc 48. Sazawal S, Black RE, Ramsan M, et al.
Iron modulation of erythropoiesis is as- Nephrol 2013;​24:​1599-616. Effects of routine prophylactic supple-
sociated with Scribble-mediated control 34. Macdougall IC, Cooper AC. Erythro- mentation with iron and folic acid on
of the erythropoietin receptor. J Exp Med poietin resistance: the role of inf lamma- admission to hospital and mortality in pre-
2018;​215:​661-79. tion and pro-inf lammatory cytokines. school children in a high malaria trans-
21. Suragani RN, Cadena SM, Cawley SM, Nephrol Dial Transplant 2002;​17:​Suppl 11:​ mission setting: community-based, ran-
et al. Transforming growth factor-β super- 39-43. domised, placebo-controlled trial. Lancet
family ligand trap ACE-536 corrects ane- 35. Kimachi M, Fukuma S, Yamazaki S, 2006;​367:​133-43.
mia by promoting late-stage erythropoie- et al. Minor elevation in C-reactive protein 49. Song S-NJ, Tomosugi N, Kawabata H,
sis. Nat Med 2014;​20:​408-14. levels predicts incidence of erythropoiesis- Ishikawa T, Nishikawa T, Yoshizaki K.
22. Stefanova D, Raychev A, Deville J, et al. stimulating agent hyporesponsiveness Down-regulation of hepcidin resulting
Hepcidin protects against lethal Esche- among hemodialysis patients. Nephron from long-term treatment with an anti-
richia coli sepsis in mice inoculated with 2015;​131:​123-30. IL-6 receptor antibody (tocilizumab) im-
isolates from septic patients. Infect Im- 36. Cavill I, Bentley DP. Erythropoiesis in proves anemia of inflammation in multi-
mun 2018;​86(7):​e00253-18. the anaemia of rheumatoid arthritis. Br J centric Castleman disease. Blood 2010;​
23. Stefanova D, Raychev A, Arezes J, Haematol 1982;​50:​583-90. 116:​3627-34.
et al. Endogenous hepcidin and its agonist 37. Mitlyng BL, Singh JA, Furne JK, Ruddy J, 50. Weiss LM, Gonzalez E, Miller SB,
mediate resistance to selected infections Levitt MD. Use of breath carbon monox- Agudelo CA. Severe anemia as the pre-
by clearing non-transferrin-bound iron. ide measurements to assess erythrocyte senting manifestation of giant cell arteri-
Blood 2017;​130:​245-57. survival in subjects with chronic diseases. tis. Arthritis Rheum 1995;​38:​434-6.
24. Truman-Rosentsvit M, Berenbaum D, Am J Hematol 2006;​81:​432-8. 51. Song S-NJ, Iwahashi M, Tomosugi N,

1156 n engl j med 381;12 nejm.org  September 19, 2019

The New England Journal of Medicine


Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Anemia of Inflammation

et al. Comparative evaluation of the effects 58. Theurl I, Schroll A, Sonnweber T, et al. Momelotinib inhibits ACVR1/ALK2, de-
of treatment with tocilizumab and TNF-α Pharmacologic inhibition of hepcidin ex- creases hepcidin production, and amelio-
inhibitors on serum hepcidin, anemia re- pression reverses anemia of chronic in- rates anemia of chronic disease in rodents.
sponse and disease activity in rheumatoid flammation in rats. Blood 2011;​118:​4977- Blood 2017;​129:​1823-30.
arthritis patients. Arthritis Res Ther 2013;​ 84. 66. Mesa RA, Kiladjian JJ, Catalano JV,
15:​R141. 59. Hohlbaum AM, Gille H, Trentmann S, et al. SIMPLIFY-1: a phase III randomized
52. Gil-Santana L, Cruz LAB, Arriaga MB, et al. Sustained plasma hepcidin suppres- trial of momelotinib versus ruxolitinib in
et al. Tuberculosis-associated anemia is sion and iron elevation by Anticalin-­ Janus kinase inhibitor-naïve patients with
linked to a distinct inflammatory profile derived hepcidin antagonist in cynomol- myelofibrosis. J Clin Oncol 2017;​35:​3844-
that persists after initiation of antituber- gus monkey. Br J Pharmacol 2018;​175:​ 50.
cular therapy. Sci Rep 2019;​9:​1381. 1054-65. 67. Peterson P, Whatcott CJ, et al. TP-0184
53. Hedenus M, Birgegård G. The role of 60. Boyce M, Warrington S, Cortezi B, inhibits ALK2/ACVR1, decreases hepcidin
iron supplementation during epoietin et al. Safety, pharmacokinetics and phar- levels, and demonstrates activity in pre-
treatment for cancer-related anemia. Med macodynamics of the anti-hepcidin Spie- clinical mouse models of functional iron
Oncol 2009;​26:​105-15. gelmer lexaptepid pegol in healthy sub- deficiency. Blood 2017;​130:​Suppl 1:​937.
54. Cody JD, Hodson EM. Recombinant jects. Br J Pharmacol 2016;​173:​1580-8. abstract.
human erythropoietin versus placebo or 61. Sheetz M, Barrington P, Callies S, et al. 68. Barrett TD, Palomino HL, Brondstet-
no treatment for the anaemia of chronic Targeting the hepcidin-ferroportin path- ter TI, et al. Prolyl hydroxylase inhibition
kidney disease in people not requiring way in anaemia of chronic kidney disease. corrects functional iron deficiency and
dialysis. Cochrane Database Syst Rev 2016;​ Br J Clin Pharmacol 2019;​85:​935-48. inflammation-induced anaemia in rats.
1:​CD003266. 62. Vadhan-Raj S, Abonour R, Goldman Br J Pharmacol 2015;​172:​4078-88.
55. Fishbane S, Ross DW, Hong S. Ane- JW, et al. A first-in-human phase 1 study 69. Chen N, Qian J, Chen J, et al. Phase 2
mia in non-dialysis-dependent CKD: to of a hepcidin monoclonal antibody, studies of oral hypoxia-inducible factor
treat or not to treat? Am J Kidney Dis LY2787106, in cancer-associated anemia. prolyl hydroxylase inhibitor FG-4592 for
2019;​73:​297-9. J Hematol Oncol 2017;​10:​73. treatment of anemia in China. Nephrol
56. Martí-Carvajal AJ, Agreda-Pérez LH, 63. Sasu BJ, Cooke KS, Arvedson TL, et al. Dial Transplant 2017;​32:​1373-86.
Solà I, Simancas-Racines D. Erythropoie- Antihepcidin antibody treatment modu- 70. Wouters HJCM, van der Klauw MM,
sis-stimulating agents for anemia in rheu- lates iron metabolism and is effective in a de Witte T, et al. Association of anemia
matoid arthritis. Cochrane Database Syst mouse model of inflammation-induced with health-related quality of life and sur-
Rev 2013;​2:​CD000332. anemia. Blood 2010;​115:​3616-24. vival: a large population-based cohort
57. Poli M, Asperti M, Ruzzenenti P, et al. 64. Kovac S, Böser P, Cui Y, et al. Anti- study. Haematologica 2019;​104:​468-76.
Oversulfated heparins with low anticoag- hemojuvelin antibody corrects anemia 71. Girelli D, Busti F. Anemia and adverse
ulant activity are strong and fast inhibi- caused by inappropriately high hepcidin outcomes in the elderly: a detrimental
tors of hepcidin expression in vitro and levels. Haematologica 2016;​101(5):​e173- inflammatory loop? Haematologica 2019;​
in vivo. Biochem Pharmacol 2014;​92:​467- e176. 104:​417-9.
75. 65. Asshoff M, Petzer V, Warr MR, et al. Copyright © 2019 Massachusetts Medical Society.

images in clinical medicine


The Journal welcomes consideration of new submissions for Images in Clinical
Medicine. Instructions for authors and procedures for submissions can be found
on the Journal’s website at NEJM.org. At the discretion of the editor, images that
are accepted for publication may appear in the print version of the Journal,
the electronic version, or both.

n engl j med 381;12 nejm.org  September 19, 2019 1157


The New England Journal of Medicine
Downloaded from nejm.org on September 18, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.

Das könnte Ihnen auch gefallen