You are on page 1of 8

JACC: HEART FAILURE VOL. 6, NO.

3, 2018

ª 2018 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION

PUBLISHED BY ELSEVIER

STATE-OF-THE-ART REVIEW

Anemia in Heart Failure


Still Relevant?

Niels Grote Beverborg, MD, Dirk J. van Veldhuisen, MD, PHD, Peter van der Meer, MD, PHD

ABSTRACT

One-third of all patients with heart failure have anemia, and its presence is associated with more symptoms, increased
rates of hospitalization, and increased mortality. The etiology of anemia is multifactorial, complex, and varies between
patients. The most important factors leading to anemia in heart failure are inadequate erythropoietin production
resulting from renal failure, intrinsic bone marrow defects, medication use, and nutritional deficiencies such as iron
deficiency. Erythropoiesis-stimulating agents (ESAs) have been proven to successfully correct hemoglobin levels, albeit
without significant improvement in clinical outcome. On the contrary, the use of ESAs has led to increased rates of
thromboembolic events and ischemic stroke. This use of ESAs for the treatment of anemia in heart failure, therefore,
cannot be recommended. In addition, these results question whether anemia is a therapeutic target or merely a
marker of disease severity. Other therapies are being studied and include agents targeting the erythropoietin receptor,
hepcidin pathway, or iron availability. This review focuses on the pathophysiology of anemia in heart failure, explains
why investigated therapies might not have led to the desired results, and discusses promising future therapies.
(J Am Coll Cardiol HF 2018;6:201–8) © 2018 by the American College of Cardiology Foundation.

I n patients with heart failure (HF), organ systems


receive, in varying degrees, an inadequate sup-
ply of oxygen and nutrients. Together with
inflammation and neurohormonal pathway activa-
management strategies? Or, in short, is anemia in pa-
tients with HF still relevant? The present review sum-
marizes the literature on anemia in patients with HF.

tion, comorbidities such as iron deficiency (50%) DIAGNOSIS AND PREVALENCE


and anemia (37%) are highly prevalent (1). Conven-
tionally, iron deficiency and anemia were considered The strict definition of anemia is an absolute decrease
cause and consequence. However, with the latest re- in red blood cell mass, which can be determined by an
sults of large trials targeted at either iron deficiency extensive and costly analysis of radiolabeled blood
or anemia, striking differences were observed in volume analysis. In clinical practice, however, only
treatment effect. Therapies aimed at raising hemoglo- derived parameters such as Hb level and hematocrit
bin (Hb) levels did not seem to be beneficial to date, are used. Hb and hematocrit are concentration
whereas treating iron deficiency resulted in substan- dependent, and in volume overloaded HF patients,
tial clinical benefits, also in HF patients without ane- hemodilution-induced “pseudo-anemia” is a recur-
mia. This outcome raises several questions: is a low rent phenomenon (2). According to the World Health
Hb level a therapeutic target? Is it merely a signal of Organization, anemia occurs at an Hb level <13 g/dl in
disease severity or an underlying comorbidity (e.g., men and <12 g/dl in women. This definition has not
renal failure, iron deficiency)? Are we using the right been validated, but in the general population with

From the Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
The University Medical Center of Groningen has received an unrestricted grant from Vifor Pharma. Dr. van Veldhuisen has
received board membership fees and travel expenses from Vifor Pharma. Dr. van der Meer has received consultancy fees from
Vifor Pharma. Dr. Grote Beverborg has reported that he has no relationships relevant to the contents of this paper to disclose.

Manuscript received May 10, 2017; revised manuscript received August 25, 2017, accepted August 31, 2017.

ISSN 2213-1779/$36.00 https://doi.org/10.1016/j.jchf.2017.08.023


202 Grote Beverborg et al. JACC: HEART FAILURE VOL. 6, NO. 3, 2018

Anemia in Heart Failure MARCH 2018:201–8

ABBREVIATIONS normal renal function, serum erythropoietin and, subsequently, aggravation of symptoms such as
AND ACRONYMS levels rise exponentially in those with Hb dyspnea and fatigue, and thus further impair exercise
levels <13 and 12 g/dl, respectively (3). In tolerance and quality of life (19).
CKD = chronic kidney disease
patients with HF, studies have reported a In a large meta-analysis with 153,180 patients with
ESA = erythropoiesis-
wide range of anemia prevalence (17% to HF, the crude mortality risk of anemia was an odds
stimulating agent
70%), which may be the result of differences ratio of 1.96 (95% confidence interval: 1.74 to 2.21),
Hb = hemoglobin
in anemia definition, patient demographic and the adjusted hazard ratio was 1.46 (95%
HF = heart failure
characteristics, comorbidities, study type confidence interval: 1.26 to 1.69), with no difference
HIF = hypoxia-inducible factor
(registry vs. trial), and HF severity (4–6). between patients with a reduced or preserved left
LVEF = left ventricular ejection
ventricular ejection fraction (LVEF) (4). In 2
fraction ETIOLOGY AND PATHOPHYSIOLOGY
observational studies, anemia resolved in 6 months’
TSAT = transferrin saturation
time in >40% of outpatients (5,6). These patients
VO2 max = maximal oxygen The etiology of anemia in patients with HF is
had a prognosis similar to those without anemia,
consumption multifactorial. Patients with concomitant
whereas persistent anemia was associated with the
chronic kidney disease (CKD) or diabetes mellitus,
poorest survival. Iron supplementation and the
higher age, and more advanced disease are at the
erythropoiesis-stimulating agent (ESA) therapy rate
highest risk of anemia (5,6). HF can cause anemia
were relatively low (21% and 8%, respectively), and
through different pathophysiological mechanisms,
resolution of anemia was hypothesized to be the ef-
and both conditions share several risk factors (Central
fect of HF treatment for a large part, particularly
Illustration). Patients with HF often have hematinic
better control of fluid status, and thus resolving
deficiencies, especially iron deficiency, which is pre-
pseudo-anemia (5). The combination of anemia, CKD,
sent in approximately one-half of the patients (7–9).
and/or iron deficiency in patients with HF often oc-
The presence of chronic inflammation in HF is an
curs, and is associated with progression of CKD and
important cause of (functional) iron deficiency and of
HF and unfavorable prognosis (20). However, it is
erythropoietin resistance (10). Inadequate levels of
unclear whether anemia leads to advanced HF and
erythropoietin, conversely, are often seen in patients
worse outcome or if anemia is merely a sign of more
with concomitant CKD because the production of
advanced disease.
erythropoietin occurs in the kidney (11). In addition,
bone marrow unresponsiveness to erythropoietin due MANAGEMENT OPTIONS
to intrinsic bone marrow defects further increases the
susceptibility to anemia (12). This action is associated TRANSFUSION THERAPY. In case of severe, symp-
with excessively elevated erythropoietin levels in tomatic anemia, blood transfusion with packed red
patients with HF and preserved erythropoietin pro- blood cells is often considered. However, data in pa-
duction; high erythropoietin levels are associated tients with HF are limited. Transfusion therapy has
with unfavorable outcome in these patients and the only temporary benefits and additional risks in pa-
development of HF in the general population (13,14). tients with HF such as volume overload and ischemic
In addition, the activated renin-angiotensin- events. Two observational studies (n ¼ 596,456 and
aldosterone system results in salt and fluid reten- n ¼ 4,102) concluded that patients with HF who
tion leading to pseudo-anemia (2,11). received blood transfusions have worse clinical fea-
Medication prescribed in HF can result in anemia. tures and prognosis, although the smaller study
Angiotensin-converting enzyme inhibitors inhibit noted that the transfusion itself seemed to be safe
hematopoietic activity via N-acetyl-seryl-aspartyl- and even beneficial compared with propensity score–
lysyl-proline, leading to a higher risk of anemia as matched patients with HF who did not receive a blood
observed in the SOLVD (Studies of Left Ventricular transfusion (21,22). Because of the risks of acute he-
Dysfunction) trial with enalapril (15,16). In addition, molytic reactions, infection, acute lung injury,
there is evidence that carvedilol might decrease Hb allergic reactions, and the lack of evidence to suggest
levels by blocking the b2-adrenergic receptor (17). a liberal transfusion strategy in patients with heart
CLINICAL CONSEQUENCES disease, a restrictive transfusion strategy (trigger
threshold of 7 to 8 g/dl) is recommended by the
In healthy individuals, oxygen delivery at Hb levels American College of Physicians (23).
as low as 5 g/dl are compensated by increases in both ERYTHROPOIESIS-STIMULATING AGENTS. Exogenous
heart rate and stroke volume, mechanisms already erythropoietin is approved for the treatment of ane-
impaired in patients with HF (18). Anemia in HF, mia as a result of CKD or chemotherapy-induced
therefore, could lead to decreased oxygen delivery anemia. In HF, the effect of anemia treatment with
JACC: HEART FAILURE VOL. 6, NO. 3, 2018 Grote Beverborg et al. 203
MARCH 2018:201–8 Anemia in Heart Failure

C ENTR AL I LL U STRA T I O N Anemia in Heart Failure: Common Ground, Cause, or Consequence?

Grote Beverborg, N. et al. J Am Coll Cardiol HF. 2018;6(3):201–8.

Anemia and heart failure share several prevalent risk factors. In addition, heart failure can lead to anemia via a large number of mechanisms, and anemia, in turn, can
lead to an increased cardiac workload and possible further deterioration of cardiac function and prognosis. ACEi ¼ angiotensin-converting enzyme inhibitor;
ASA ¼ acetylsalicylic acid.

ESAs on outcome was examined in the RED-HF thrombotic events (153 [13.5%] vs. 114 [10.0%];
(Reduction of Events by Darbepoetin Alfa in Heart p ¼ 0.009) were increased in those treated with dar-
Failure) trial, which, with 2,278 patients, is the largest bepoetin alfa (24,25). This outcome led to further
study to date (24). In this study, patients with safety concerns because increased rates of ischemic
symptomatic chronic HF (LVEF #40%) and anemia stroke and thrombotic events with ESAs were also
(Hb level 9.0 to 12.0 g/dl) were randomized to receive observed in patients with CKD and chemotherapy-
darbepoetin alfa (with a target of 13 to 14.5 g/dl) or induced anemia (26,27). Possible reasons for these
placebo. Co-treatment with oral or intravenous iron results include the wrong therapy, wrong target
was allowed in both groups. Median Hb levels in the population, or wrong target Hb level.
intervention group increased, but no effect was As noted earlier, the etiology of anemia in HF is
observed on the primary composite endpoint of death very heterogeneous (7–12,28). Patients with a trans-
or hospitalization for worsening HF or any of the ferrin saturation (TSAT) <15% were excluded from
other endpoints. To the contrary, rates of ischemic the RED-HF trial, but this method still leaves
stroke (41 [4.5%] vs. 32 [2.8%]; p ¼ 0.03) and embolic/ patients with iron deficiency with TSAT 15% to 20%
204 Grote Beverborg et al. JACC: HEART FAILURE VOL. 6, NO. 3, 2018

Anemia in Heart Failure MARCH 2018:201–8

and a ferritin level 100 to 300 m g/l or a ferritin Hepcidin is the regulator of iron metabolism and is
level <100 mg/l included in the trial. In addition, no involved in the pathophysiology of the anemia of
other investigations into the cause of anemia were chronic disease. The hormone is upregulated in
performed. The presence of pseudo-anemia has been inflammation and degrades the iron exporter ferro-
proposed as a possible reason for the neutral results, portin, thus blocking iron uptake from the gut and
which was also reported in a small study of 28 iron release from macrophages. This process provides
anemic patients treated with erythropoietin alfa (29). a possible explanation for the neutral results of the
Because a large portion of patients with HF already IRONOUT study.
have a disproportionately high erythropoietin level Intravenous iron has been studied in 5 randomized
associated with bone marrow resistance to erythro- clinical trials (Table 1) (32–35). All studies included
poietin, administering even more erythropoietin to patients based on their ferritin and TSAT levels; Toblli
these patients would be counterintuitive (13). This et al. (32) and Anker et al. (34) additionally used a
approach is supported by data from the RED-HF trial, relatively low Hb level as an inclusion criterion (<12.5
which showed that approximately one-quarter of and <13.5 g/dl, respectively). Despite differences in
patients with HF did not exhibit any increase in Hb treatment strategies and follow-up, the overall re-
level after 4 weeks of ESA treatment, and this unre- sults of the trials were broadly similar: treatment with
sponsiveness to ESAs is independently associated intravenous iron led to improvements in New York
with hospitalizations and all-cause mortality (30). Heart Association functional class, exercise capacity,
Regarding the Hb target, no data comparing different and quality of life in a short period of time. In 2 of the
targets in patients with HF are available, but data largest trials, FAIR-HF (Ferinject Assessment in
from trials in CKD indicate that higher Hb targets may Patients with Iron Deficiency and Chronic Heart
result in worse cardiovascular outcome (27). Failure) and CONFIRM-HF (Ferric Carboxymaltose
Evaluation on Performance in Patients With Iron
IRON THERAPY. Iron therapy was initially adminis- Deficiency in Combination With Chronic Heart Fail-
tered as co-therapy in trials with ESAs, mainly as oral ure), significant overall increases in Hb levels were
therapy (24). More recently, the awareness of the high observed, but the treatment effect was similar
prevalence of iron deficiency (w70% in the anemic in anemic and nonanemic patients (34,35). The
and w50% of the entire HF population), the clinical third study is the EFFECT-HF (Effect of Ferric
consequences of iron deficiency, and the availability Carboxymaltose on Exercise Capacity in Patients
of new intravenous iron formulations have led to the With Iron Deficiency and Chronic Heart Failure), a
design of trials investigating iron therapy without randomized controlled trial of intravenous ferric car-
ESAs. The initial trials were performed in patients boxymaltose compared with standard of care in 172
with anemia but later also in patients with iron iron-deficient patients with HF (36). Primary analysis
deficiency irrespective of the presence of anemia. showed an increase in VO 2 max in patients treated
Although oral iron has practical advantages over with ferric carboxymaltose compared with a control
intravenous iron, its use in HF seems limited because group that was not treated. In addition, an increase in
of therapy compliance issues due to gastrointestinal the Hb level of 0.74  0.17 g/dl was reported after
side effects and impaired iron uptake. The latter was 24 weeks. However, subgroup analyses of anemic and
also observed in the only randomized placebo- nonanemic patients are not yet available.
controlled Phase II study with oral iron, the IRON- In summary, although anemia and iron deficiency
OUT (Oral Iron Repletion Effects On Oxygen Uptake exhibit a large overlap, isolated iron deficiency is
in Heart Failure) (31). This study included 225 HF prevalent, and the benefits of treating iron deficiency
patients with a reduced LVEF and iron deficiency. probably extend beyond hematopoiesis. The effects
Patients received 150 mg of polysaccharide iron of intravenous iron on hard clinical endpoints remain
complex or placebo, twice daily for 16 weeks. Only a to be established. Given these results, it is recom-
marginal increase of 11 mg/l of ferritin and 3% TSAT mended to screen for iron deficiency in all patients
with oral iron was observed, with no significant effect with HF, independent of their Hb level. To diagnose
on exercise capacity (measured according to maximal iron deficiency, the combination of TSAT and ferritin
oxygen consumption [VO 2 max]) or N-terminal pro–B- levels (ferritin <100 m g/l or a ferritin level 100 to
type natriuretic peptide level. In exploratory ana- 300 mg/l with a TSAT<20%) has most often been used
lyses, changes in TSAT correlated with changes in VO2 in large clinical trials. However, this definition has not
max and N-terminal pro–B-type natriuretic peptide been validated, and ferritin levels are often unreliable
levels. The only patients who responded to oral iron because it is an acute-phase reactant. We recently
therapy were those with low hepcidin levels. presented data from a study in which we validated the
JACC: HEART FAILURE VOL. 6, NO. 3, 2018 Grote Beverborg et al. 205
MARCH 2018:201–8 Anemia in Heart Failure

T A B L E 1 Randomized Controlled Trials With Intravenous Iron in Patients With Heart Failure

First Author, Definition of Study


Year (Ref. #) N Population Iron Deficiency Therapy Period Effect on Hb Results

Toblli et al., 40 Hb <12.5 g/dl, Ferritin <100 mg/l Iron sucrose 200 mg 26 weeks From 10.3  0.6 g/dl at NT-proBNP Y
2007 (32) LVEF #35%, þ TSAT <20% every 5 weeks baseline to 11.8  0.7 CRP Y
eGFR <90 ml/min g/dl at 6 months NYHA functional
(p < 0.01) in the class Y
intervention group; no LVEF [
significant difference in eGFR [
control group 6MWT [
MLHFQ [
Okonko et al., 35* NYHA functional class Ferritin <100 mg/l or Iron sucrose 200 mg 16 weeks 0.1 (–0.8 to 0.9) g/dl at VO2 max [
FERRIC-HF, II–III, LVEF #45%, VO2 ferritin 100–300 mg/l weekly till ferritin 16 weeks (p ¼ 0.87) (p ¼ 0.08)
2008 (33) max <18 ml/kg/min, þ TSAT <20% >500 mg/l† VO2 max/kg [
Hb <14.5 g/dl NYHA functional
class Y
PGA [
Anker et al., FAIR- 459* NYHA functional class Ferritin <100 mg/l or FCM 200 mg until 24 weeks FCM vs. placebo NYHA functional
HF, 2009 (34) II–III, LVEF #40%, ferritin 100–299 mg/l normalized iron 13.0  1 g/dl vs. 12.5  1 class Y
Hb 9.5–13.5 g/dl þ TSAT <20% status‡ g/dl (p < 0.001) at PGA [
24 weeks 6MWT [
EQ-5D [
KCCQ [
Ponikowski et al., 304 NYHA functional class Ferritin <100 mg/l or FCM 500–2,000 mg at 52 weeks 0.6  0.2 g/dl and 1.0  6MWT [
CONFIRM-HF, II–III, LVEF #45%, ferritin 100–300 mg/l baseline and after 6 0.2 g/dl after 24 NYHA functional
2015 (35) Hb <15 g/dl, þ TSAT <20% weeks; subsequently, and 52 weeks (both class Y
NT-proBNP >400 500 mg every 12 p < 0.001) PGA [
pg/ml, or BNP weeks if still iron EQ-5D [
>100 pg/ml deficient HF hospitalizations Y
(not a
predefined
endpoint)
van Veldhuisen 172 NYHA functional class Ferritin <100 mg/l or FCM 500–2,000 mg at 24 weeks 0.74  0.17 g/dl after VO2 max [
et al., EFFECT- II–III, LVEF #45%, ferritin 100–300 mg/l baseline and after 6 24 weeks (p < 0.0001) NYHA functional
HF, 2017 (36) VO2 max 10–20 þ TSAT <20% and 12 weeks if still class Y
ml/kg/min, Hb <15 g/ iron deficient PGA [
dl, NT-proBNP >400
pg/ml, or BNP >100
pg/ml

*2:1 randomization, 24 (FERRIC-HF [Ferric Iron Sucrose in Heart Failure]) and 304 (FAIR-HF [Ferinject Assessment in Patients with Iron Deficiency and Chronic Heart Failure]) patients in the treatment group.
†Ferritin >500 mg/l; subsequently, 200 mg once a month. ‡Calculated by using the Ganzoni formula; after iron normalization, 200 mg once every 4 weeks.
6MWT ¼ 6-min walking test; BNP ¼ brain natriuretic peptide; CONFIRM-HF ¼ Ferric Carboxymaltose Evaluation on Performance in Patients With Iron Deficiency in Combination With Chronic Heart Failure;
CRP ¼ C-reactive protein; EFFECT-HF ¼ Effect of Ferric Carboxymaltose on Exercise Capacity in Patients With Iron Deficiency and Chronic Heart Failure; eGFR ¼ estimated glomerular filtration rate; EQ-5D ¼
5-dimension European Quality of Life; FCM ¼ ferric carboxymaltose; Hb ¼ hemoglobin; HF ¼ heart failure; KCCQ ¼ Kansas City Cardiomyopathy Questionnaire; LVEF ¼ left ventricular ejection fraction;
MLHFQ ¼ Minnesota Living with Heart Failure Questionnaire; NYHA ¼ New York Heart Association; NT-proBNP ¼ N-terminal pro–B-type natriuretic peptide; PGA ¼ patient global assessment; TSAT ¼
transferrin saturation; VO2 max ¼ maximum oxygen uptake per minute.

cutoff of TSAT<20% by using the gold standard of bone focus on determining the underlying etiology and
marrow iron staining (37). Ferritin had no diagnostic subsequent treatment, although, often no specific
value. We supported these findings by showing that a cause is found. Special attention is paid to iron defi-
low TSAT, and not a low ferritin level, was associated ciency and its treatment with intravenous ferric car-
with an increased risk of mortality. This finding is in boxymaltose. The use of the ESA darbepoetin-alfa
line with results from the recently published meta- is not recommended by the European Society of
analysis of Anker et al. (38) who found that, using Cardiology (39).
interaction analysis, patients with a TSAT $20.1% do
not respond to iron therapy with improved outcomes, AREAS OF DEVELOPMENT
whereas those in the lower tertiles of TSAT do respond.
Several different strategies are currently being
CURRENT GUIDELINE RECOMMENDATIONS explored for the treatment of anemia in HF. These
strategies are aimed directly at the process of eryth-
The most recent guidelines of the American College of ropoiesis by targeting the erythropoietin receptor or
Cardiology Foundation/American Heart Association hypoxia pathway but also indirectly via the hepcidin
and the European Society of Cardiology both recog- pathway (Figure 1).
nize anemia as an important comorbidity in patients HEPCIDIN. Hepcidin, the master iron regulator, can
with HF (39,40). Management recommendations be antagonized in several ways: 1) decreasing
206 Grote Beverborg et al. JACC: HEART FAILURE VOL. 6, NO. 3, 2018

Anemia in Heart Failure MARCH 2018:201–8

F I G U R E 1 New Therapies for Anemia and Their Targets in Erythropoiesis

Production
HIF-stabilizers
BMP inhibitors
Anti-cytokines
siRNAs

Neutralizing

mAb
EPO Activin traps Hepcidin Spiegelmers
Anticalins

Ferroportin Binding

EPO mimetics Anti-ferroportin


GDF11+ cells
rhEPO antibodies
Iron

+ + + +
Efficacy

Pluripotent stem cell BFU-E CFU-E Proerythroblasts Erythroblasts Reticulocytes RBCs

ErythropoieƟn dependent phase Iron dependent phase

The process of production of new red blood cells (RBCs): erythropoiesis. The first stages are dependent on erythropoietin (EPO). During the erythroblasts stage, iron
availability is essential as it is incorporated in hemoglobin. Most new therapies target either EPO or iron. Hypoxia-inducible factor (HIF) stabilizers affect both
pathways. Although not fully understood, the data suggest that activin receptor ligand traps also addresses both pathways and increases efficacy of erythropoiesis by
reducing the number of growth differentiation factor (GDF)-11-positive cells. Hepcidin can be antagonized by decreasing hepcidin production, neutralizing hepcidin, or
preventing hepcidin–ferroportin interaction. As a result of hepcidin inhibition, ferroportin expression is increased, and iron absorption and iron availability for
erythropoiesis increase. BFU-E ¼ erythroid burst-forming units; BMP ¼ bone morphogenic protein; CFU-E ¼ erythroid colony-forming units; mAb ¼ monoclonal
antibodies; rhEPO ¼ recombinant human erythropoietin; siRNA ¼ small interfering ribonucleic acid.

hepcidin production; 2) neutralizing hepcidin; or 3) fusion proteins, gene therapy, and mimetic peptides)
preventing hepcidin–ferroportin interaction. Agents and activin receptor ligand traps. Activin traps are
neutralizing hepcidin are the most promising to date. recombinant fusion proteins consisting of the
One phase I study showed that a fully humanized immunoglobulin G1 Fc domain linked to the extra-
monoclonal antibody against hepcidin (LY2787106) cellular domain of the activin receptor IIA. These bind
was well tolerated and resulted in increases in serum a number of transforming growth factor-b superfam-
iron levels and TSAT in patients with cancer and ily ligands, including activin A and activin B, and
anemia (41). Another hepcidin-binding agent, the thereby inhibit their signaling. Sotatercept, the most
Spiegelmer lexaptepid (NOX-H94), has been shown to studied activin trap, was initially studied as an oste-
increase serum iron levels in healthy persons sub- oporosis agent. Surprisingly, an increase in Hb level,
jected to inflammation by injection of lipopolysac- red blood cell number, and hematocrit were observed
charides (42). A small Phase II study found an Hb (43). Together with the prevention of vascular calci-
increase of $1 g/dl after 4 weeks of treatment with fication shown in a small trial with 43 patients with
lexaptepid in 5 of 12 patients with functional iron- end-stage renal disease, this drug might prove bene-
deficiency anemia (30). Currently, the results of ficial for the elderly CKD population (44). Animal
trials investigating the effects of these agents in ESA models point to growth differentiation factor 11 as the
hyporesponsive, anemic patients undergoing dialysis target for these drugs, which is a differentiator in-
are awaited. hibitor present on erythroid progenitors (45). How-
ever, increased expression of angiotensin II has also
ERYTHROPOIETIN RECEPTOR TARGETING. Drugs been proposed as one of the possible mechanisms of
directly targeting erythropoiesis include erythropoi- increased erythropoiesis by stimulating erythroid
etin receptor–targeting drugs (receptor antibodies, differentiation directly through the AT1 receptor or
JACC: HEART FAILURE VOL. 6, NO. 3, 2018 Grote Beverborg et al. 207
MARCH 2018:201–8 Anemia in Heart Failure

via induction of erythropoietin production by the ancillary properties may have unknown side effects,
kidney (46). Increased angiotensin II levels are clearly and there is fear of serious adverse events such as
not desirable in an HF population and thus far, no promotion of tumor growth. Future studies are needed
studies in HF have been conducted. to address safety and efficacy.

CONCLUSIONS
HYPOXIA-INDUCIBLE FACTOR STABILIZERS. The
final and most promising drugs are the hypoxia-
Anemia in patients with HF is still relevant even
inducible factor (HIF) stabilizers. HIF is the master
though the therapies studied thus far have not shown
regulator of the cellular response to hypoxia (47). It is
positive clinical results. It is present in approximately
rapidly degraded in the presence of oxygen, but in
one-third of patients with HF, and these patients
low oxygen conditions, it induces transcription of
have a worse prognosis and poor quality of life.
>60 genes (including erythropoietin and vascular
Anemia may indicate several underlying conditions,
endothelial growth factor). HIF stabilizers can be
such as nutritional deficiencies, renal disease, and
administered orally and induce physiological eryth-
volume overload, although in the latter case, this
ropoietin levels. Several Phase I and II studies in pa-
condition is called pseudo-anemia. Despite the
tients with CKD have shown promising results with
consensus that underlying disorders should be
increasing levels of Hb and decreasing levels of hep-
addressed, there is no evidence for the clinical benefit
cidin after therapy (48). One of the compounds with
of increasing Hb levels as such. ESA therapy has
the most data available is FG-4592, or roxadustat.
shown neutral results on rates of death and HF
Roxadustat was recently shown to be effective in
rehospitalization and leads to more ischemic strokes,
increasing Hb levels and to be superior compared
which outweigh their marginal effect on symptom
with epoetin alfa in correcting anemia in patients
improvement. Intravenous iron therapy looks prom-
with CKD in 2 different phase II trials (49). One study
ising for iron-deficiency anemia, but its benefit is
comprised 87 patients undergoing dialysis who
partly independent from Hb levels, and data on hard
received different doses of roxadustat or placebo, and
clinical endpoints are not yet available.
the other study included 91 patients who were not yet
dialysis-dependent who continued with epoetin alfa or ACKNOWLEDGMENT The authors acknowledge M.A.
switched to different doses of roxadustat. Although Kooij, BSc, for assistance with the illustrations.
only oral iron supplementation was allowed, iron
indices remained stable or increased, and reduced ADDRESS FOR CORRESPONDENCE: Dr. Peter van
hepcidin levels were observed in both studies. How- der Meer, Department of Cardiology, University
ever, caution is warranted: because HIF stabilizers Medical Center Groningen, Hanzeplein 1, PO Box
affect many biological pathways (including fatty acid 30.001, 9700 RB Groningen, the Netherlands.
and glucose metabolism, as well as angiogenesis), such E-mail: p.van.der.meer@umcg.nl.

REFERENCES

1. van Veldhuisen DJ, Anker SD, Ponikowski P, ambulatory patients with chronic heart failure. 10. van der Meer P, Lok DJ, Januzzi JL, et al.
Macdougall IC. Anemia and iron deficiency in heart J Am Coll Cardiol 2008;51:569–76. Adequacy of endogenous erythropoietin levels
failure: mechanisms and therapeutic approaches. and mortality in anaemic heart failure patients. Eur
6. Waldum B, Westheim AS, Sandvik L, et al.
Nat Rev Cardiol 2011;8:485–93. Heart J 2008;29:1510–5.
Baseline anemia is not a predictor of all-cause
2. Adlbrecht C, Kommata S, Hülsmann M, et al. mortality in outpatients with advanced heart fail- 11. Westenbrink BD, Visser FW, Voors AA, et al.
Chronic heart failure leads to an expanded plasma ure or severe renal dysfunction. Results from the Anaemia in chronic heart failure is not only related
volume and pseudoanaemia, but does not lead to Norwegian Heart Failure Registry. J Am Coll Car- to impaired renal perfusion and blunted erythro-
a reduction in the body’s red cell volume. Eur diol 2012;59:371–8. poietin production, but to fluid retention as well.
Heart J 2008;29:2343–50. Eur Heart J 2007;28:166–71.
7. Klip IT, Comin-Colet J, Voors AA, et al. Iron
3. Grote Beverborg N, Verweij N, Klip IT, et al. deficiency in chronic heart failure: an international 12. Westenbrink BD, Voors AA, De Boer RA, et al.
Erythropoietin in the general population: refer- pooled analysis. Am Heart J 2013;165:575–82.e3. Bone marrow dysfunction in chronic heart failure
ence ranges and clinical, biochemical and genetic patients. Eur J Heart Fail 2010;12:676–84.
8. Okonko DO, Mandal AK, Missouris CG, Poole-
correlates. PLoS One 2015;10:e0125215.
Wilson PA. Disordered iron homeostasis in chronic 13. van der Meer P, Voors AA, Lipsic E, Smilde TDJ,
4. Groenveld HF, Januzzi JL, Damman K, et al. heart failure: prevalence, predictors, and relation van Gilst WH, van Veldhuisen DJ. Prognostic value
Anemia and mortality in heart failure patients. a to anemia, exercise capacity, and survival. J Am of plasma erythropoietin on mortality in patients
systematic review and meta-analysis. J Am Coll Coll Cardiol 2011;58:1241–51. with chronic heart failure. J Am Coll Cardiol 2004;
Cardiol 2008;52:818–27. 44:63–7.
9. Nanas JN, Matsouka C, Karageorgopoulos D,
5. Tang WH, Tong W, Jain A, Francis GS, Harris CM, et al. Etiology of anemia in patients with 14. Grote Beverborg N, van der Wal HH, Klip IT,
Young JB. Evaluation and long-term prognosis of advanced heart failure. J Am Coll Cardiol 2006; et al. High serum erythropoietin levels are related
new-onset, transient, and persistent anemia in 48:2485–9. to heart failure development in subjects from the
208 Grote Beverborg et al. JACC: HEART FAILURE VOL. 6, NO. 3, 2018

Anemia in Heart Failure MARCH 2018:201–8

general population with albuminuria: data from treatment of cancer-associated anemia. JAMA 38. Anker SD, Kirwan BA, van Veldhuisen DJ, et al.
PREVEND. Eur J Heart Fail 2016;18:814–21. 2008;299:914. Effects of ferric carboxymaltose on hospital-
isations and mortality rates in iron-deficient heart
15. van der Meer P, Lipsic E, Westenbrink BD, et al. 27. Singh AK, Szczech L, Tang KL, et al. Correction
failure patients: an individual patient data meta-
Levels of hematopoiesis inhibitor N-acetyl-seryl- of anemia with epoetin alfa in chronic kidney dis-
analysis. Eur J Heart Fail 2018;20:125–33.
aspartyl-lysyl-proline partially explain the occur- ease. N Engl J Med 2006;355:2085–98.
rence of anemia in heart failure. Circulation 2005; 39. Ponikowski P, Voors AA, Anker SD, et al. 2016
28. Kleijn L, Belonje AM, Voors AA, et al.
112:1743–7. ESC guidelines for the diagnosis and treatment of
Inflammation and anaemia in a broad spectrum
16. Ishani A, Weinhandl E, Zhao Z, et al. acute and chronic heart failure. Eur Heart J 2016;
of patients with heart failure. Heart 2012;98:
Angiotensin-converting enzyme inhibitor as a risk 37:2129–200.
1237–41.
factor for the development of anemia, and the 40. Yancy CW, Jessup M, Bozkurt B, et al. 2013
29. Borovka M, Teruya S, Alvarez J, Helmke S,
impact of incident anemia on mortality in patients ACCF/AHA guideline for the management of heart
Maurer MS. Differences in blood volume compo-
with left ventricular dysfunction. J Am Coll Cardiol failure: a report of the American College of Car-
nents between hyporesponders and responders to
2005;45:391–9. diology Foundation/American Heart Association
erythropoietin alfa: the Heart Failure With Pre-
17. Komajda M, Anker SD, Charlesworth A, et al. Task Force on Practice Guidelines. J Am Coll Car-
served Ejection Fraction (HFPEF) anemia trial.
The impact of new onset anaemia on morbidity diol 2013;62:e147–239.
J Card Fail 2013;19:685–91.
and mortality in chronic heart failure: results from 41. Vadhan-Raj S, Abonour R, Goldman JW, et al.
30. van Veldhuisen DJ, Pfeffer MA, van der
COMET. Eur Heart J 2006;27:1440–6. A first-in-human phase 1 study of a hepcidin
Meer P, Olson K, Solomon SD. Abstract 1293:
18. Weiskopf RB, Viele MK, Feiner J, et al. Human hyporesponsiveness to darbepoetin alfa in pa- monoclonal antibody, LY2787106, in cancer-
cardiovascular and metabolic response to acute, tients with heart failure and anemia in the RED-HF associated anemia. J Hematol Oncol 2017;10:73.
severe isovolemic anemia. JAMA 1998;279:217–21. study: clinical and prognostic associations. Eur J 42. van Eijk LT, John AS, Schwoebel F, et al. Effect
19. Ebner N, Jankowska EA, Ponikowski P, et al. Heart Fail 2015;17:269. of the antihepcidin Spiegelmer lexaptepid on
The impact of iron deficiency and anaemia on ex- 31. Lewis GD, Semigran MJ, Givertz MM, et al. Oral inflammation-induced decrease in serum iron in
ercise capacity and outcomes in patients with iron therapy for heart failure with reduced ejection humans. Blood 2014;124:2643–6.
chronic heart failure. Results from the studies fraction: design and rationale for oral iron reple- 43. Ruckle J, Jacobs M, Kramer W, et al. Single-
investigating co-morbidities aggravating heart tion effects on oxygen uptake in heart failure. Circ dose, randomized, double-blind, placebo-
failure. Int J Cardiol 2016;205:6–12. Heart Fail 2016;9:e000345. controlled study of ACE-011 (ActRIIA-IgG1) in
20. Klip IT, Jankowska EA, Enjuanes C, et al. 32. Toblli JE, Lombraña A, Duarte P, Di Gennaro F. postmenopausal women. J Bone Miner Res 2009;
The additive burden of iron deficiency in the Intravenous iron reduces NT-pro-brain natriuretic 24:744–52.
cardiorenal-anaemia axis: scope of a problem and peptide in anemic patients with chronic heart 44. Havill J, Kopyt N, Coyne D, Weiswasser M,
its consequences. Eur J Heart Fail 2014;16: failure and renal insufficiency. J Am Coll Cardiol Smith W. Sotatercept improves anemia,
655–62. 2007;50:1657–65. vascular calcification, and bone loss in patients
21. Garty M, Cohen E, Zuchenko A, et al. Blood with end-stage kidney disease on hemodialysis
33. Okonko DO, Grzeslo A, Witkowski T, et al. Ef-
transfusion for acute decompensated heart fail- (Abstr. TH-PO038). J Am Soc Nephrol 2015;26:
fect of intravenous iron sucrose on exercise
ure—friend or foe? Am Heart J 2009;158:653–8. 310A–1A.
tolerance in anemic and nonanemic patients with
22. Kao DP, Kreso E, Fonarow GC, Krantz MJ. symptomatic chronic heart failure and iron defi- 45. Paulson RF. Targeting a new regulator of
Characteristics and outcomes among heart failure ciency FERRIC-HF: a randomized, controlled, erythropoiesis to alleviate anemia. Nat Med 2014;
patients with anemia and renal insufficiency with observer-blinded trial. J Am Coll Cardiol 2008;51: 20:334–5.
and without blood transfusions (public discharge 103–12.
46. Iancu-Rubin C, Mosoyan G, Wang J,
data from California 2000-2006). Am J Cardiol 34. Anker SD, Comin Colet J, Filippatos G, et al. Kraus T, Sung V, Hoffman R. Stromal cell-
2011;107:69–73. Ferric carboxymaltose in patients with heart fail- mediated inhibition of erythropoiesis can be
23. Qaseem A, Humphrey LL, Fitterman N, ure and iron deficiency. N Engl J Med 2009;361: attenuated by Sotatercept (ACE-011), an activin
Starkey M, Shekelle P, Clinical Guidelines Com- 2436–48. receptor type II ligand trap. Exp Hematol 2013;
mittee of the American College of Physicians. 41:155–66.e17.
35. Ponikowski P, Van Veldhuisen DJ, Comin-
Treatment of anemia in patients with heart dis- Colet J, et al. Beneficial effects of long-term 47. Semenza GL. Hypoxia-inducible factor 1:
ease: a clinical practice guideline from the Amer- intravenous iron therapy with ferric carbox- master regulator of O2 homeostasis. Curr Opin
ican College of Physicians. Ann Intern Med 2013; ymaltose in patients with symptomatic heart fail- Genet Dev 1998;8:588–94.
159:770–9. ure and iron deficiency. Eur Heart J 2015;36: 48. Bonomini M, Del Vecchio L, Sirolli V,
24. Swedberg K, Young JB, Anand IS, et al. 657–68. Locatelli F. New treatment approaches for the
Treatment of anemia with darbepoetin alfa in anemia of CKD. Am J Kidney Dis 2016;67:133–42.
36. van Veldhuisen DJ, Ponikowski P, van der
systolic heart failure. N Engl J Med 2013;368:
Meer P, et al. Effect of ferric carboxymaltose on 49. Chen N, Qian J, Chen J, et al. Phase 2
1210–9.
exercise capacity in patients with chronic heart studies of oral hypoxia-inducible factor prolyl
25. Bello NA, Lewis EF, Desai AS, et al. Increased failure and iron deficiency. Circulation 2017;136: hydroxylase inhibitor FG-4592 for treatment of
risk of stroke with darbepoetin alfa in anaemic 1374–83. anemia in China. Nephrol Dial Transplant 2017;
heart failure patients with diabetes and chronic 32:1373–86.
37. Grote Beverborg N, van der Meer P. Definition
kidney disease. Eur J Heart Fail 2015;17:1201–7.
of iron deficiency based on the gold standard of
26. Bennett CL. Venous thromboembolism and bone marrow iron staining and treatment effect of
mortality associated with recombinant erythro- ferric carboxymaltose in heart failure patients KEY WORDS anemia, erythropoiesis-
poietin and darbepoetin administration for the (abstr). Eur J Heart Fail 2017;19:20–1. stimulating agents, heart failure, iron

You might also like