Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Canadian Journal of Cardiology - (2015) 1e9

Systematic Review/Meta-analysis
The Efficacy and Safety of Iron Supplementation in Patients
With Heart Failure and Iron Deficiency: A Systematic Review
and Meta-analysis
Cheng Qian, MD,* Baozhu Wei, MD, PhD,* Jinye Ding, MD, Huiting Wu, MD,
and Yanggan Wang, MD, PhD
Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China

ABSTRACT 
RESUM 
E
Background: Iron deficiency (ID) is a common comorbidity in patients Introduction : Une carence en fer (CF) est une comorbidite  fre
quente
with heart failure (HF) and has been associated with increased mor- chez les patients atteints d’insuffisance cardiaque (IC) et elle est
tality and hospitalizations. However, the benefit and safety of iron associe e à une augmentation de la mortalite  et des hospitalisations.
supplementation in treating HF and ID in randomized controlled trials Toutefois, le be ne
fice et la se
curite
 d’une supple
mentation en fer dans
(RCTs) are inconclusive. We therefore performed a meta-analysis to le traitement de l’IC et de la CF dans des essais contrôles randomise s
overcome this limitation. (ECR) ne sont pas concluants. Nous avons donc re alise
 une me ta-
Methods: PubMed, the Cochrane Library, and ClinicalTrials.gov were analyse pour surmonter cette limitation.
systematically searched for eligible trials up to December 31, 2014. Me thodes : PubMed, la Cochrane Library, et le site ClinicalTrials.gov
We also searched the references of all relevant studies and reviews for ont e te
 syste
matiquement fouille s pour des essais admissibles jus-
more trials. Only RCTs reporting the clinical impact of iron therapy in qu’au 31 de cembre 2014. Nous avons e galement cherche  les
patients with HF with ID compared with no iron treatment were  fe
re rences de toutes les e tudes et commentaires pertinents pour plus
enrolled in our meta-analysis. d’essais. Seuls les ECR rapportant l’impact clinique d’un apport

Iron deficiency (ID) is now viewed as a frequent and impor- aerobic metabolism and oxidative phosphorylation.8 Cells
tant comorbidity in patients with heart failure (HF). Studies with high-energy demand, such as cardiomyocytes and skeletal
have reported a high prevalence of ID in patients with chronic myocytes, are particularly sensitive to ID. As the impaired
HF of up to 30%-50%.1,2 Traditionally, the clinically related energy generation and utilization in the myocardium and the
influences of ID were considered only in the context of ane- peripheral tissue are always involved in the pathophysiological
mia. However, ID anemia can be regarded as the end phase of process of HF,9 correction of ID may be an efficient treatment
a process beginning with the gradual depletion of iron stores.3 for HF and ID. In the past few decades, a number of trials
Accordingly, even in patients with a preserved hemoglobin have explored the effect of erythropoiesis-stimulating agents
level, ID may already exist in chronic HF.4 ID, with or (ESAs) and iron preparations in treating patients with chronic
without anemia, is associated with exercise intolerance, low HF. Treatment with ESAs alone failed to reduce mortality or
quality of life, and increased mortality and hospitalization in hospitalization for HF while it increased the risk of throm-
patients with HF.5-7 boembolic events.10 ESAs are thus not recommended to treat
Besides improving erythropoiesis, intravenous iron has anemic patients with HF.11 ESAs combined with iron have
been shown to maintain energy metabolism by augmenting been shown to improve cardiac function, New York Heart
Association (NYHA) functional class, and exercise
capacity.12-14 Two observational studies, together with 1 pre-
Received for publication April 6, 2015. Accepted June 16, 2015. post trial, have found that monotherapy with intravenous iron
*These authors contributed equally to this work. improved symptoms, cardiac remodelling and exercise
Corresponding author: Dr Yanggan Wang, Department of Cardiology, intolerance.15-17 In addition, iron therapy alone has shown to
Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang have the same benefits as the combination of ESAs and iron in
District, Wuhan 430071, Hubei Province, China. Tel.: þ86-27-67812844;
fax: þ86-027-67812844.
patients with HF.18 Although almost all randomized
E-mail: wb000813@whu.edu.cn controlled trials (RCTs) have observed that intravenous iron
See page 8 for disclosure information. could improve the exercise capacity, functional status, and

http://dx.doi.org/10.1016/j.cjca.2015.06.009
0828-282X/Ó 2015 Canadian Cardiovascular Society. Published by Elsevier Inc. All rights reserved.
2 Canadian Journal of Cardiology
Volume - 2015

Results: Five clinical trials comprising a total of 907 patients were therapeutique de fer pour des patients en IC avec CF, comparative-
finally included. Compared with placebo or no treatment, additional ment à aucun apport en fer, ont e  te
 inclus dans notre me ta-analyse.
iron therapy was associated with a significantly reduced rate of hos- Resultats : Cinq essais cliniques comprenant un total de 907 patients
pitalization for HF (odds ratio [OR], 0.28; 95% confidence interval [CI], ont finalement e te
 inclus. Comparativement au placebo ou aucun
0.16-0.49), though all-cause mortality was not significantly different traitement, une therapie supplemente e en fer a ete
 associe e à un taux
(OR, 0.81; 95% CI, 0.42-1.57). In 4 studies where these endpoints significativement re duit d’hospitalisation en cas d’IC (ratio d’incidence
were combined, the incidence of hospitalization for HF and death was approche  [RIA], 0,28; intervalle de confiance [IC] à 95 %, de 0,16 à
lowered in the iron supplementation group (OR, 0.47; 95% CI, 0.29- 0,49), bien que la mortalite  toutes causes confondues n’e tait pas sig-
0.76). There was no increase in the risk of adverse events. nificativement diffe rente (RIA, 0,81; IC à 95 %, 0,42 à 1,57). Dans
Conclusions: Iron supplementation significantly reduced the risk of (a) quatre etudes où ces points limites ont e te combine s, l’incidence des
hospitalization for HF and (b) the combined endpoint of hospitalization hospitalisations pour IC et de la mortaliteetait diminuee dans le groupe
for HF and death, without increasing the risk of adverse events in avec une supple mentation en fer (RIA, 0,47; IC à 95 %, 0,29 à 0,76). Il
patients with symptomatic systolic HF and ID. However, the current n’y avait pas d’augmentation du risque d’e venements inde sirables.
data are inadequate to make a clear determination upon mortality. Conclusions : La supple mentation en fer re duit significativement le
risque (a) d’une hospitalisation pour IC et (b) du critère combine  des
hospitalisations pour IC et de la mort, sans augmenter le risque d’ef-
fets indesirables chez les patients pre sentant des symptômes d’IC
systolique et de CF. Cependant, les donne es actuelles sont insuffi-
santes pour obtenir un avis tranche  concernant la mortalite .

quality of life in iron-deficient patients with HF, whether it [OR]) were also recorded for the pooled analysis. All dis-
has benefit in terms of reducing mortality or hospitalization crepancies between the two reviewers were resolved by dis-
remains unknown. Individual RCTs also have not been large cussion with a third reviewer (J.D.).
enough to detect the prognostic effects of iron supplementa-
tion. Therefore, a meta-analysis pooling the data from RCTs
is needed to overcome this limitation. Quality assessment and study outcome
The quality of the included articles was assessed according
Methods to Cochrane collaboration’s tool for assessing the risk of
bias.20 The efficacy endpoints for this analysis included hos-
Data sources and search strategy pitalization for HF, all-cause mortality, and a combined
endpoint of hospitalization for HF and death, whereas adverse
This study was conducted according to Preferred Report-
events (AEs) were recorded as the safety endpoint. We
ing Items for Systematic Reviews and Meta-Analyses state-
collected the quantitative data on only severe AEs reported in
ment.19 We performed systematic literature searches in the
each RCT for synthesis, because their concrete definition was
database of PubMed, the Cochrane Library, and
stated only in 1 eligible study.21
ClinicalTrials.gov for eligible studies. The reference lists of
relevant studies and reviews were also checked to find addi-
tional trials. We combined the following search terms: “iron”
or “ferrous” or “ferric,” and “heart failure” or “cardiac failure”
or “myocardial failure” or “heart decompensation.” Our
search was limited to human and English articles published
until December 31, 2014.

Inclusion criteria
Clinical studies that met the following requirements were
included in our meta-analysis: (a) RCTs; (b) enrolled patients
with established HF and ID regardless of left ventricular
ejection fraction (LVEF); (c) compared iron therapy with no
iron therapy (placebo or no treatment); (d) reported data
about hospitalization for HF, all-cause mortality, a combined
endpoint of hospitalization for HF and death, and adverse
events. Trials that compared the combined treatment of ESAs
and iron with no iron treatment were excluded.

Data extraction
Two reviewers (C.Q. and B.W.) independently extracted
the details regarding study and patient characteristics, treat-
ment strategy, ID definition, and duration of follow-up. The
total number of clinical endpoints and effect sizes (odds ratio Figure 1. Flow diagram for inclusion of studies in this meta-analysis.
Qian et al. 3
Iron Supplementation for HF and Iron Deficiency

Statistical analysis

ACEI, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; BB, b-blocker; FCM, ferric carboxymaltose; Hb, hemoglobin; HF, heart failure; ID, iron deficiency; IS, iron sucrose;
Follow-up
(wk)
24

18

14

24

52
Data analysis was performed using STATA 12.0 (Stata-
Corp, College Station, TX). ORs with 95% confidence in-
tervals (CIs) for all outcomes were used to present the pooled
effect size of the individual studies. Statistical heterogeneity

Ferritin < 100 ng/mL or TSAT

300 ng/mL if TSAT < 20%)

300 ng/mL if TSAT < 20%)

300 ng/mL if TSAT < 20%)


Ferritin < 100 ng/mL (or 100-

Ferritin < 100 ng/mL (or 100-

Ferritin < 100 ng/mL (or 100-


across the studies was quantified using the I2 statistic. When I2
> 25% (indicate significant heterogeneity), we reported a

Definition of ID
pooled estimate based on the random-effects model; otherwise
the fixed-effects model was used. Sensitivity analysis was
conducted to assess the robustness of the pooled estimates by
(a) switching to the other effects model and (b) removing each

< 20%
trial one at a time from the analysis and evaluating the overall
estimates for the remaining studies. The results of the pooled

NS
effect size were considered to be statistically significant only at
P < 0.05.

Diuretic
95

71

92

90
Results

Medication (%)
ACEI/ARB
Study selection and baseline characteristics

NS
98/23

74/20

78/24
The study selection process is displayed in Fig. 1. Briefly,

IV, intravenous; LVEF, left ventricular ejection fraction; NS, not stated; NYHA, New York Heart Association; TSAT, transferrin saturation.
92z
of the initial 1512 citations, 327 were excluded because 165
studies were non-English articles, and 162 reports were not

100

89

85

90
BB
human trials. After scanning the titles and abstracts, we
excluded 1152 studies irrelevant to our participant. The
remaining 33 studies and 1 additional trial (identified by
Ischemic
HF (%)

reviewing the reference list of a study22) were selected for full-


62.5

74.3

80.2

83.4
NS
text screening, during which 29 publications did not meet the
inclusion criteria. Consequently, 5 RCTs21,23-26 performed
between 2007 and 2014 were finally included in our meta-
Hb (g/dL)

9.5-13.5
analysis.
< 11.5y
< 12.5

< 12.5

< 15
Table 1 summarizes the baseline characteristics of the
Inclusion criteria

eligible studies. All of them were designed using the ran-


domized controlled method and compared additional intra-
NYHA

NS

venous iron treatment with placebo or no treatment. Ferric


II-IV

II-III

II-III

II-III

carboxymaltose24-26 and iron sucrose21,23,24 were used in 3


separate studies (1 trial24 used both) with the dose varying
 45%*

from 200 to 1000 mg. A total of 907 participants with


 45%

 45%

 40%

 45%
LVEF

chronic HF were included, of whom 556 were assigned to the


iron group and 351 to the control group. The average age of
the participants in each study ranged from 62 to 76 years, and
Male

58.6% were women. The mean duration of follow-up ranged


47.7
45.2
(%)
NS
NS

NS

NS
71
73

33

26

from 14 to 52 weeks. All patients in the included trials had


Table 1. Baseline characteristics of the included studies

established chronic HF with an LVEF  45% and NYHA


class II-III (except one study23 that enrolled patients with
 14
 11

68  10
67  11
69  10
Age (y)
7
8

70  9

*  40% for NYHA II and  45% for NYHA III.


NS

NS

NYHA class IV). In the included studies, the hemoglobin cut-


Percentage of patients receiving ACEI or ARB.
76
74
64
62

off value varied from 11.5 to 15.0 g/dL. ID was defined as


ferritin < 100 ng/mL or transferrin saturation (TSAT) <
< 12.5 for men and < 11.5 for women.
20
20
24
11
27
30
15
304
155
150

151

20% in 1 study,23 and 3 trials21,25,26 diagnosed ID with


N

ferritin < 100 ng/mL or between 100 and 300 ng/mL if


TSAT < 20%. In addition, 2 studies23,24 enrolled patients
Iron and control

IV FCM 200 mg

IV FCM 200 mg

IV FCM 500 or

with chronic renal failure, but the mean creatinine clearance


IV IS 200 mg

IV IS 200 mg

IV IS 200 mg
No treatment

No treatment

was greater than 30 mL/min.


1000 mg

Quality assessment
Saline

Saline

Saline

The Cochrane risk of bias assessment was used to perform


quality assessment (shown in Fig. 2). Overall, random
Ponikowski
Arutyunov
200723

200821

200924

200925

201426

sequence generation was observed in 4 studies, and 2 of them


Okonko
Toblli

Anker

had reported allocation concealment. Three trials were double


Study

y
z

blinded and had performed a blinded assessment of the


4 Canadian Journal of Cardiology
Volume - 2015

Figure 2. Risk of bias assessment in the included studies.

outcomes. Neither incomplete outcome data nor selective between iron and non-iron groups (OR, 0.81; 95% CI, 0.42-
reporting was present in any of the included studies. We also 1.57; P ¼ .53) (Fig. 3B).
did not observe other significant bias during quality assess-
ment. However, there are no data available for appraising the Hospitalization for HF and death
risk of bias in 1 study.24
Four studies reported the composite of hospitalization for
HF and death, and no proof of heterogeneity was found (I2 ¼
Hospitalization for HF
0%, P ¼ 0.574). Overall, 31 (6.2%) patients reached the
Four included studies reported the risk of hospitalization combined endpoint in the iron group (n ¼ 499), whereas 49
for HF, and no evidence of heterogeneity was observed across (14.6%) patients reached the endpoint in the control group
them (I2 ¼ 0%, P ¼ 0.723). In total, there were 18 (3.6%) (n ¼ 336, Supplemental Table S1). Pooling across the 4 trials
hospitalizations due to HF in the iron group (n ¼ 499), using fixed-effect meta-analysis indicated that iron supple-
compared with 47 (14.0%) hospitalizations in the control mentation was associated with a significant decrease in the risk
group (n ¼ 336, Supplemental Table S1). The pooled esti- of this combined endpoint (OR, 0.47; 95% CI, 0.29-0.76;
mates using fixed-effect meta-analysis showed a significantly P ¼ 0.002) (Fig. 4).
reduced rate of hospitalization for HF among the iron sup-
plementation patients compared with the non-iron treatment Adverse events
group (OR, 0.28; 95% CI, 0.16-0.49; P < 0.001) (Fig. 3A).
Data for severe AEs were provided in 4 studies, and sta-
tistical heterogeneity across them was not significant (I2 ¼
All-cause mortality
19%, P ¼ 0.295). In summary, there were 52 (9.7%) par-
Four studies reported data for all-cause mortality, and there ticipants with at least one severe AE in the iron group (n ¼
was no significant heterogeneity across them (I2 ¼ 0%, P ¼ 538), whereas 62 (18.7%) participants in the control group
0.962). In general, 19 (3.5%) patients in the iron group (n ¼ had at least one severe AE (n ¼ 332, Supplemental Table S2).
536) reached this endpoint compared with 18 (5.4%) patients Meta-analytic pooling for this safety outcome using a fixed-
in the control group (n ¼ 331, Supplemental Table S1). effects model revealed that the iron group had a statistically
Pooling across the 4 trials using fixed-effect meta-analysis lower rate of severe AEs than the control group (OR, 0.50;
showed no significant difference in all-cause mortality 95% CI, 0.34-0.75; P ¼ 0.001) (Fig. 5). Of the AEs reported
Qian et al. 5
Iron Supplementation for HF and Iron Deficiency

Figure 3. Forest plot showing the effect of iron supplementation on (A) hospitalization for HF and (B) all-cause mortality in patients with HF and ID.

in the included studies, such as cardiac, nervous system, Discussion


gastrointestinal, and vascular disorders, none was observed to In the present meta-analysis, we summarize the available
occur more frequently in the iron group. In addition, no se- evidence on the prognostic effect and safety of iron treatment
vere allergic reactions related to the iron treatment were re- in patients with HF and ID. The results demonstrate that iron
ported in each included trial. supplementation is associated with a significant reduction in
(a) hospitalization because of HF and (b) the combined
endpoint of hospitalization for HF and death. However, we
failed to detect the superiority in terms of all-cause mortality
Sensitivity analysis of iron therapy to non-iron treatment or placebo. Addition-
When switching to random-effects model analysis, the ally, iron supplementation is not associated with an increased
overall ORs are consistent with the direction and size of the risk of adverse events in patients with chronic HF.
ones calculated by the fixed-effects model. Elimination of each Similar benefits of iron therapy were also observed in 2
trial one at a time from the analysis did not have any relevant previous meta-analyses.27,28 Although our study additionally
influence on the overall results with the exception of the effect included only one large and well-designed RCT published
variable regarding severe AEs, which confirmed in direction recently, the sample size of our meta-analysis increased by
and magnitude the major results of the present study and about half. Moreover, the most recent meta-analysis enrolled
suggested that there was only a nominal reduction in the risk patients were treated in combination with ESAs and lost the
of severe AEs in this meta-analysis (shown in Table 2). significance of prognostic endpoints after exclusion of some
6 Canadian Journal of Cardiology
Volume - 2015

Figure 4. Forest plot showing the effect of iron supplementation on the combined outcome of hospitalization for HF and death in patients with HF
and ID.

trials.28 Conversely, we conducted a reliable sensitivity anal- iron therapy has been shown to improve quality of life and
ysis and evaluated more endpoints (eg, the combined out- exercise tolerance.11 In Ferinject Assessment in Patients With
comes of death and HF hospitalization). The above points Iron Deficiency and Chronic Heart Failure (FAIR-HF)25 and
increase accuracy and robustness of our findings. Ferric Carboxymaltose Evaluation on Performance in Patients
Several studies have confirmed the negative effects of ID on With Iron Deficiency in Combination With Chronic Heart
aerobic capacity, endurance, energetic efficiency, and work Failure (CONFIRM-HF)26 studies, significant improvements
productivity.29 According to the experimental data, iron were observed with intravenous iron preparation for 6-minute
treatment improves the impaired electron transport of walk distance, quality-of-life assessment, and NYHA func-
myocardial mitochondria and exercise performance in iron- tional class. Moreover, hospitalizations for worsening HF are
deficient rats,30,31 indicating its role as an important always related to unfavorable clinical outcomes. Therefore,
cofactor in energy metabolism. In addition, long-term iron- our finding that additional iron use significantly reduced the
deficiency anemia can induce cardiac maladaptation, risk of hospitalization for HF, consistent with the results of
including cardiac dilatation and myocardial fibrosis.32 For the the CONFIRM-HF26 study, is physiologically and clinically
patients with chronic HF and ID (with or without anemia), credible.

Figure 5. Forest plot showing the impact of iron supplementation on severe adverse events (defined as deaths, all-cause hospitalizations, and life-
threatening events) in patients with HF and ID.
Qian et al. 7
Iron Supplementation for HF and Iron Deficiency

Table 2. Sensitivity analysis


Hospitalization for HFy All-cause mortalityy Combined endpointy Severe adverse eventsy
Study* or
effect model OR (95% CI) P OR (95% CI) P OR (95% CI) P OR (95% CI) P
Toblli23 0.30 (0.17-0.54) < 0.001 NA NA 0.52 (0.31-0.85) 0.009 NA NA
Okonko21 0.27 (0.15-0.49) < 0.001 0.79 (0.40-1.55) 0.49 0.45 (0.28-0.74) 0.002 0.50 (0.33-0.74) < 0.001
Arutyunov24 NA NA 0.81 (0.41-1.59) 0.54 NA NA 0.50 (0.33-0.75) < 0.001
Anker25 0.24 (0.12-0.49) < 0.001 0.88 (0.41-1.87) 0.73 0.44 (0.25-0.79) 0.005 0.75 (0.42-1.35) 0.34
Ponikowski26 0.29 (0.12-0.70) 0.006 0.73 (0.23-1.88) 0.59 0.43 (0.20-0.92) 0.03 0.37 (0.22-0.63) < 0.001
Random 0.29 (0.16-0.52) < 0.001 0.81 (0.42-1.56) 0.52 0.49 (0.30-0.80) 0.004 0.51 (0.31-0.85) 0.009
CI, confidence interval; HF, heart failure; NA, not assessed; OR, odds ratio.
* Removing each study (author’s name) one at a time.
y
Evaluating the overall estimate of each endpoint for the remaining studies or random-effects model.

There is a trend towards lower mortality in iron-treated ferritin < 100 ng/mL (absolute ID) or TSAT < 20% in one
patients although not reaching statistical significance, as included study. But when ferritin is between 100 and 300 ng/
shown in the present analysis. Nevertheless, it should be mL, the diagnosis of ID is complex and only functional ID
mentioned that we observed that only 4.3% of the overall can be diagnosed (with TSAT < 20%).37 Definitions con-
cohort experienced this outcome that is mainly due to the cerning absolute and functional ID should thus be combined,
small sample size and short follow-up duration in all of the which has been applied in 3 late trials we included. Agents for
included trials, which may be inadequate to detect potential treating HF including b-blockers, angiotensin-converting
differences. To evaluate the composite prognostic effects, we enzyme inhibitors and/or angiotensin receptor blockers, and
pooled the combined endpoint of hospitalization for HF and diuretics were widely used in the included trials. The high
death and found that iron supplementation was related to a proportions of patients receiving these recommended HF
significant reduction in the risk of this composite outcome. medications, comparable to what was seen in large HF trials,
The positive finding further confirmed the prognostic benefit were not significantly different across the studies, suggesting
of additional iron therapy in patients with chronic HF their critical role in the management of HF. Nevertheless,
and ID. because of the negative prognostic value of ID, it is insuffi-
We did not observe that intravenous iron supplementation cient that only using these agents to treat patients with HF
was associated with an increased risk of AEs, including car- with the comorbidity. Therefore, intravenous iron should be
diovascular, nervous system, and gastrointestinal disorders. considered for optimal treatment of patients with HF who are
Conversely, intravenous iron seems to result in fewer severe iron deficient.
AEs, but this statistically significant benefit was not well Although advances in the management of HF have made
established because of vague results in the sensitivity analysis. great contributions to improving the outcomes of systolic HF,
The FAIR-HF25 and CONFIRM-HF26 studies are 2 large- postdischarge mortality and readmission rates in patients with
scale RCTs that may drive the results of our meta-analysis. HF remain unacceptably high.38 Moreover, the rate of reho-
Both of them showed benefit in terms of a reduction in HF spitalization continues to rise and approach 30% within 60 to
hospitalizations, or approximately the same magnitude. 90 days of discharge.39 What’s worse is that the mean cost of
However, by removing each one of them in sensitivity anal- HF-related hospitalization has imposed a huge economic
ysis, we did not find statistically substantial changes in overall burden on health care resources.40 Intravenous iron prepara-
results. The etiology of HF does not seem to be an important tion, such as ferric carboxymaltose, has been estimated to be
confounder for the pooled estimates, because the high pro- cost-effective in treating patients with HF and ID.41 There-
portions (approximately 65%-80%) of patients with ischemic fore, the results of our meta-analysis showing that iron sup-
causes of HF are similar between iron and control groups in plementation was associated with a significant reduction in
all included trials. Moreover, in the 2 large-scale RCTs,25,26 a hospitalization because of HF are of particular clinical
consistent benefit regarding the 6-minute walk distance was significance.
observed in patients with HF with and without ischemic There are some limitations in our meta-analysis. First, we
causes. did not consider the influences of the iron preparation dose,
There are extremely limited data about the efficacy and the hemoglobin level, and the NYHA class on therapeutic
safety of oral iron in treating HF. Despite being a relatively effects because of the absence of relevant data. Second,
cheap preparation, oral iron is poorly absorbed, particularly in because only 2 trials administered ferric carboxymaltose, 2
patients with chronic diseases, such as chronic HF, and is not trials administered iron sucrose, and 1 trial administered both
well tolerated because of adverse gastrointestinal effects.33,34 without some relevant data, analyzing the class-specific effects
In a recent study with a small sample size, improvement in of iron preparations on clinical outcomes is inappropriate.
VO2max was found with intravenous iron but not oral iron.35 Third, the sample size and follow-up duration in these clinical
In addition, a meta-analysis indicates that intravenous iron is trials were small and short, respectively, resulting in a lack of
indeed more effective than oral iron in chronic kidney dis- statistical power; hence, a larger sample size and a longer
ease.36 Thus, intravenous iron may be superior to the oral iron duration of follow-up may be needed to observe significant
in terms of clinical efficacy and safety. changes in prognostic endpoints. Forth, it must be mentioned
The therapy for patients with HF with ID should also be that all included trials enrolled patients with symptomatic HF
optimized. Regarding this issue, a proper definition of ID is of with an LVEF below 45%, so generalization of our findings
great importance. As mentioned above, ID was defined as should be used with caution.
8 Canadian Journal of Cardiology
Volume - 2015

Conclusions function and functional cardiac class, and markedly reduces hospitaliza-
Based on the results of our meta-analysis, iron supple- tions. J Am Coll Cardiol 2000;35:1737-44.
mentation significantly reduced risk of (a) rehospitalizations 13. Silverberg DS, Wexler D, Sheps D, et al. The effect of correction of mild
because of HF and (b) the combined endpoint of rehospi- anemia in severe, resistant congestive heart failure using subcutaneous
talizations for HF and death, without increased adverse events erythropoietin and intravenous iron: a randomized controlled study.
in patients with symptomatic systolic HF and ID. However, J Am Coll Cardiol 2001;37:1775-80.
the current data are inadequate to make a conclusive deter-
14. Comin-Colet J, Ruiz S, Cladellas M, Rizzo M, Torres A, Bruguera J.
mination upon total mortality. Additional well-designed and A pilot evaluation of the long-term effect of combined therapy with
large RCTs with long-term follow-up duration are certainly intravenous iron sucrose and erythropoietin in elderly patients with
needed to confirm our findings especially regarding mortality, advanced chronic heart failure and cardio-renal anemia syndrome: in-
in terms of the effects of different iron preparations and doses, fluence on neurohormonal activation and clinical outcomes. J Card Fail
different hemoglobin levels and NYHA classes, and different 2009;15:727-35.
baseline LVEF values.
15. Usmanov RI, Zueva EB, Silverberg DS, Shaked M. Intravenous iron
without erythropoietin for the treatment of iron deficiency anemia in
Funding Sources patients with moderate to severe congestive heart failure and chronic
kidney insufficiency. J Nephrol 2008;21:236-42.
This work was supported by the National Natural Science
Foundation of China (Grant no. 81270304). 16. Bolger AP, Bartlett FR, Penston HS, et al. Intravenous iron alone for the
treatment of anemia in patients with chronic heart failure. J Am Coll
Cardiol 2006;48:1225-7.
Disclosures
The authors have no conflicts of interest to disclose. 17. Gaber R, Kotb NA, Ghazy M, Nagy HM, Salama M, Elhendy A. Tissue
Doppler and strain rate imaging detect improvement of myocardial
function in iron deficient patients with congestive heart failure after iron
References replacement therapy. Echocardiography 2012;29:13-8.
1. Jankowska EA, Rozentryt P, Witkowska A, et al. Iron deficiency: an 18. Terrovitis JV, Kaldara E, Ntalianis A, et al. Intravenous iron alone is
ominous sign in patients with systolic chronic heart failure. Eur Heart J equally effective with the combination of iron and erythropoietin for the
2010;31:1872-80. treatment of iron-deficiency anemia in advanced heart failure. J Am Coll
Cardiol 2012;60:2255-6.
2. Klip IT, Comin-Colet J, Voors AA, et al. Iron deficiency in chronic heart
failure: an international pooled analysis. Am Heart J 2013;165: 19. Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA Group. Preferred
575-582.e3. reporting items for systematic reviews and meta-analyses: the PRISMA
statement. BMJ 2009;339:b2535.
3. Beard JL. Iron biology in immune function, muscle metabolism and
neuronal functioning. J Nutr 2001;131:568S-79S [discussion: 580S]. 20. Higgins JPT, Green S. Cochrane Collaboration. Cochrane Handbook for
Systematic Reviews of Interventions. Chichester, England/Hoboken, NJ:
4. Besarab A, Horl WH, Silverberg D. Iron metabolism, iron deficiency,
Wiley-Blackwell, 2008xxi. 649 pp.
thrombocytosis, and the cardiorenal anemia syndrome. Oncologist
2009;14(Suppl 1):22-33. 21. Okonko DO, Grzeslo A, Witkowski T, et al. Effect of intravenous iron
sucrose on exercise tolerance in anemic and nonanemic patients with
5. Jankowska EA, Rozentryt P, Witkowska A, et al. Iron deficiency predicts
symptomatic chronic heart failure and iron deficiency FERRIC-HF: a
impaired exercise capacity in patients with systolic chronic heart failure.
randomized, controlled, observer-blinded trial. J Am Coll Cardiol
J Card Fail 2011;17:899-906.
2008;51:103-12.
6. Enjuanes C, Klip IT, Bruguera J, et al. Iron deficiency and health-related
22. Anker SD, Colet JC, Filippatos G, et al. Rationale and design of Ferinject
quality of life in chronic heart failure: results from a multicenter European
assessment in patients with IRon deficiency and chronic Heart Failure
study. Int J Cardiol 2014;174:268-75.
(FAIR-HF) study: a randomized, placebo-controlled study of intravenous
7. Rangel I, Goncalves A, de Sousa C, et al. Iron deficiency status irre- iron supplementation in patients with and without anaemia. Eur J Heart
spective of anemia: a predictor of unfavorable outcome in chronic heart Fail 2009;11:1084-91.
failure patients. Cardiology 2014;128:320-6. 23. Toblli JE, Lombrana A, Duarte P, Di Gennaro F. Intravenous iron re-
8. Dunn LL, Suryo Rahmanto Y, Richardson DR. Iron uptake and meta- duces NT-pro-brain natriuretic peptide in anemic patients with chronic
bolism in the new millennium. Trends Cell Biol 2007;17:93-100. heart failure and renal insufficiency. J Am Coll Cardiol 2007;50:
1657-65.
9. Ingwall JS. Energy metabolism in heart failure and remodelling. Car-
diovasc Res 2009;81:412-9. 24. Arutyunov G, Bylova N, Ivleva A, Kobalava Z. The safety of intravenous
(IV) ferric carboxymaltose versus IV iron sucrose in patients with chronic
10. Swedberg K, Young JB, Anand IS, et al. Treatment of anemia with heart failure (CHF) and chronic kidney disease (CKD) with iron defi-
darbepoetin alfa in systolic heart failure. N Engl J Med 2013;368:1210-9. ciency (ID). Eur J Heart Fail Suppl 2009;8. Abstract 141.

11. Moe GW, Ezekowitz JA, O’Meara E, et al. The 2014 Canadian Car- 25. Anker SD, Comin Colet J, Filippatos G, et al. Ferric carboxymaltose in
diovascular Society Heart Failure Management Guidelines Focus Update: patients with heart failure and iron deficiency. N Engl J Med 2009;361:
anemia, biomarkers, and recent therapeutic trial implications. Can J 2436-48.
Cardiol 2015;31:3-16.
26. Ponikowski P, van Veldhuisen DJ, Comin-Colet J, et al. Beneficial effects
12. Silverberg DS, Wexler D, Blum M, et al. The use of subcutaneous of long-term intravenous iron therapy with ferric carboxymaltose in pa-
erythropoietin and intravenous iron for the treatment of the anemia of tients with symptomatic heart failure and iron deficiency. Eur Heart J
severe, resistant congestive heart failure improves cardiac and renal 2015;36:657-68.
Qian et al. 9
Iron Supplementation for HF and Iron Deficiency

27. Avni T, Leibovici L, Gafter-Gvili A. Iron supplementation for the 36. Rozen-Zvi B, Gafter-Gvili A, Paul M, Leibovici L, Shpilberg O,
treatment of chronic heart failure and iron deficiency: systematic review Gafter U. Intravenous versus oral iron supplementation for the treatment
and meta-analysis. Eur J Heart Fail 2012;14:423-9. of anemia in CKD: systematic review and meta-analysis. Am J Kidney
Dis 2008;52:897-906.
28. Kapoor M, Schleinitz MD, Gemignani A, Wu WC. Outcomes of pa-
tients with chronic heart failure and iron deficiency treated with intra-
37. Jankowska EA, von Haehling S, Anker SD, Macdougall IC,
venous iron: a meta-analysis. Cardiovasc Hematol Disord Drug Targets
Ponikowski P. Iron deficiency and heart failure: diagnostic dilemmas and
2013;13:35-44.
therapeutic perspectives. Eur Heart J 2013;34:816-29.
29. Haas JD, Brownlie T. Iron deficiency and reduced work capacity: a
critical review of the research to determine a causal relationship. J Nutr 38. Ambrosy AP, Fonarow GC, Butler J, et al. The global health and eco-
2001;131:676S-88S [discussion: 688S-90S]. nomic burden of hospitalizations for heart failure: lessons learned from
hospitalized heart failure registries. J Am Coll Cardiol 2014;63:1123-33.
30. Blayney L, Bailey-Wood R, Jacobs A, Henderson A, Muir J. The effects
of iron deficiency on the respiratory function and cytochrome content of
39. Gheorghiade M, Pang PS. Acute heart failure syndromes. J Am Coll
rat heart mitochondria. Circ Res 1976;39:744-8. Cardiol 2009;53:557-73.
31. Willis WT, Gohil K, Brooks GA, Dallman PR. Iron deficiency: improved
exercise performance within 15 hours of iron treatment in rats. J Nutr 40. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for
1990;120:909-16. the management of heart failure: a report of the American College of
Cardiology Foundation/American Heart Association Task Force on
32. Naito Y, Tsujino T, Matsumoto M, Sakoda T, Ohyanagi M, Practice Guidelines. J Am Coll Cardiol 2013;62:e147-239.
Masuyama T. Adaptive response of the heart to long-term anemia
induced by iron deficiency. Am J Physiol Heart Circ Physiol 2009;296: 41. Hofmarcher T, Borg S. Cost-effectiveness analysis of ferric carbox-
H585-93. ymaltose in iron-deficient patients with chronic heart failure in Sweden.
33. McDonagh T, Macdougall IC. Iron therapy for the treatment of iron J Med Econ 2015;18:492-501.
deficiency in chronic heart failure: intravenous or oral? Eur J Heart Fail
2015;17:248-62.
34. Horl WH. Clinical aspects of iron use in the anemia of kidney disease. Supplementary Material
J Am Soc Nephrol 2007;18:382-93. To access the supplementary material accompanying this
35. Beck-da-Silva L, Piardi D, Soder S, et al. IRON-HF study: a randomized article, visit the online version of the Canadian Journal of
trial to assess the effects of iron in heart failure patients with anemia. Int J Cardiology at www.onlinecjc.ca and at http://dx.doi.org/10.
Cardiol 2013;168:3439-42. 1016/j.cjca.2015.06.009.

You might also like