• Nie Znaleziono Wyników

Effects of erythropoiesis-stimulating agents on heart failure patients with anemia: a meta-analysis

N/A
N/A
Protected

Academic year: 2022

Share "Effects of erythropoiesis-stimulating agents on heart failure patients with anemia: a meta-analysis"

Copied!
7
0
0

Pełen tekst

(1)

Corresponding author:

Hengliang Zhang MD, PhD, Department of Cardiology, The First Affiliated Hospital of Henan University of Science and Technology, Jinghua St, 471000 Luoyang, China, fax: +86 13937959952, e-mail: 271674835@qq.com

Received: 27.10.2015, accepted: 21.12.2015.

Effects of erythropoiesis-stimulating agents on heart failure patients with anemia: a meta-analysis

Hengliang Zhang

1

, Pei Zhang

1

, Yaheng Zhang

1

, Junqiang Yan

1

, Pingshuan Dong

1

, Yanyu Wang

1

, Xiaowei Niu

2

1 Department of Cardiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China

2The First College of Clinical Medical, Lanzhou University, Lanzhou, China

Adv Interv Cardiol 2016; 12, 3 (45): 247–253 DOI: 10.5114/aic.2016.61647

A b s t r a c t

Introduction: Heart failure (HF) is always complicated with anemia and is associated with bad prognosis in this patient popu- lation. Several studies have assessed the potential role of erythropoietin-stimulating agent (ESA) in improving cardiac function and reducing the number of hospitalizations in anemic patients with HF.

Aim: We performed a meta-analysis to assess the potential role of ESA in the treatment of anemic patients with HF.

Material and methods: A literature and Medline search was performed to identify studies with control groups that examined the efficacy of ESA therapy in patients with HF and anemia.

Results: A total of 11 studies were included (n = 3044 subjects) in the final analysis. Compared to placebo, ESA therapy was associated with increased hemoglobin levels (1.89 g/dl; 95% CI: 1.64–2.14, p < 0.00001), increased left ventricular ejection fraction (LVEF) to 6.88 (95% CI: 0.49–13.28, p = 0.03), decreased B-type natriuretic protein (–272.20; 95% CI: (–444.52)–(–99.89), p = 0.002), improvement in New York Heart Association functional class to –0.33 mean difference (95% CI: (–0.44)–(–0.23), p < 0.00001), and decreased hospitalization (OR = 0.61, 95% CI: 0.39–0.94, p = 0.02). There was no significant between-group difference in all-cause mortality (OR = 0.78, 95% CI: 0.51–1.21, p = 0.27).

Conclusions: The treatment of anemia with ESA therapy did not reduce the rate of all-cause mortality among patients with heart failure, but ESA therapy made a potential important contribution to patients’ symptomatic improvement.

Key words: meta-analysis, heart failure, randomized controlled trials, anemia, erythropoietin-stimulating agents.

Introduction

Heart failure (HF) is always complicated with anemia and is associated with bad prognosis in this patient pop- ulation. Depending on the definition used and specific pa- tient population studied, the prevalence rate of anemia in HF patients widely varies from 9% to 70% [1–4]. In a meta- analysis of HF patients, presence of anemia almost dou- bled the mortality risk [5]. Anemia in HF patients not only causes a higher mortality rate, but is also associated with higher rate of various morbidities such as increased num- ber of hospitalizations [6], worse New York Heart Associa- tion (NYHA) functional class [7], worse exercise capacity [8], cognitive impairment [9], and reduced quality of life [10].

The safety of erythropoiesis-stimulating agent (ESA) therapy in patients with renal failure and malignancies is not confirmed, but this is an effective therapy of ane- mia in chronic kidney disease (CKD) and cancer [11, 12].

Erythropoiesis-stimulating agent therapy can improve

the health-related quality of life (HRQL) and fatigue of patients with CKD and cancer, and this improvement is both statistically and clinically significant [13]. Several studies have explored the potential therapeutic effects of ESA in improving heart function and reducing the hos- pitalization rate in patients with HF. Other studies, how- ever, have shown that HF patients have higher levels of erythropoietin, which may lead to higher mortality [14].

Aim

We performed a meta-analysis to assess the potential role of ESA in the treatment of anemic patients with HF.

Material and methods

Search strategy

The electronic databases MEDLINE, EMBASE, the Co-

chrane Central Registry of Controlled Trials, and the Web

of Science were searched from inception to July 2015.

(2)

This search included the following terms: “heart failure”,

“congestive heart failure”, “chronic heart failure”, “CHF”,

“darbepoetin”, “erythropoiesis”, “stimulating proteins”,

“erythropoiesis stimulating protein”, “recombinant eryth- ropoietin”, “EPO/erythropoietin” and “randomized con- trolled trials”. The search was limited to English language articles of studies in adult humans. We also hand-searched potentially relevant studies, investigated registers of ongo- ing trials, and contacted the lead authors if necessary.

Study selection criteria

Two investigators (H.L.Z. and P.Z.) searched and eval- uated all titles, abstracts and full articles independently using predesigned inclusion and exclusion criteria. Any uncertainties were resolved by consensus or consulta- tion with the third investigator (J.Q.Y.) if necessary, which was infrequent. We excluded studies that 1) concerned non-clinical research; 2) included subjects without HF;

3) included subjects without anemia; 4) did not involve the administration of erythropoietin or darbepoetin; 5) in-

cluded patients less than 18 years old; 6) were published in abstract form only; 7) were not published in English.

Statistical analysis

The study was conducted following Cochrane Col- laboration meta-analysis review methodology, and data analysis was performed with the RevMan 5.3 and STATA 12.0 software package. Continuous variables with normal distribution are presented as mean ± standard deviation (SD). When studies did not directly supply the SD of the mean for the calculation of effect size, it was manually calculated from the standard error (SE) or the 95% con- fidence interval (CI). Treatment effects for continuous variables were evaluated as weighted mean difference (WMD). We pooled the results from the individual stud- ies and performed tests of heterogeneity between studies using the c

2

test and quantified by the I

2

statistic. When significant heterogeneity was detected between studies, the random effects model was used. Possible publication bias was estimated with a  funnel plot and Egger’s test.

Meta-regression analyses were performed to explore the potential sources of significant heterogeneity. To reduce the risk of over-fitting of the regression model, a minimum of nine studies was set to identify each influential factor.

Results

The primary electronic search yielded 398 studies.

After screening the titles and abstracts, 20 articles were retrieved for full-text assessment. According to the in- clusion criteria, 9 articles were excluded and a  total of 11 studies [15–25] were included (Figure 1). All of those studies were published in full-text form.

Study characteristics

This meta-analysis include 11 studies comprising 3044 patients, and the characteristics of those studies Figure 1. Process of study selection. Flowchart

shows the literature search process and the final number of studies included

Studies included in this meta-analysis (n = 11)

Articles excluded after screening titles or abstracts (n = 378)

Uncontrolled trials (n = 4) Duplicates (n = 2)

Other (n = 3)

Table I. Characteristics of included studies

Study (first

author, year) N Age [years] Anemia definition,

hemoglobin level [g/dl]

ESA therapy Follow-up

[months]

ESA Control ESA Control

Cleland, 2005 24 69 74 < 12.5 DA Placebo 2

Ghali, 2008 319 68 69 9–12 DA + iron Placebo + iron 12

Kaurea, 2008 41 73 65 < 12.5 DA + iron Placebo + iron 3

Mancini, 2003 23 87 63 Hematocrit < 35% EPO + iron Placebo + iron 3

Palazzuoli, 2006 38 72 75 < 11 EPO + iron Placebo + iron 9

Palazzuoli, 2007 51 74 72 < 11.5 EPO + iron Placebo + iron 8

Parissis, 2008 32 72 69 < 12.5 DA + iron Placebo + iron 3

Ponikowski, 2007 41 70 72 9–12 DA + iron Placebo + iron 6

Silverberg, 2001 32 75 72 10–11.5 EPO + iron Iron 8

Swedberg, 2013 2278 71 72 9–12 DA + iron Placebo + iron 28

Veldhuisen, 2007 165 71 71 9–12 DA + iron Placebo + iron 6

EPO – erythropoietin, DA – darbepoetin alfa, ESA – erythropoiesis-stimulating agent.

Potentially relevant citations identified from literature search (n = 398)

Potentially relevant full-text artickles evaluated (n = 20)

(3)

are summarized in Table I. Of those patients, 1564 re- ceived ESA therapy and 1480 were in the control group.

Baseline characteristics of the two study groups were well balanced and no significant differences were re- ported. All of the participants had typical symptoms of heart failure with left ventricular ejection fraction (LVEF)

< 40%. The baseline hemoglobin level ranged from 9.0 to 12.5 g/dl. The type of ESA therapy is darbepoetin alfa or erythropoietin. The dose regimen and target hemoglobin level were variable.

Therapeutic effects of erythropoiesis- stimulating agent

Nine studies provided data on the effect of ESA ther- apy on hemoglobin levels with an average increase of 1.89 g/dl (95% CI: 1.64–2.14, p < 0.00001) compared to

placebo (Figure 2 A). Five studies comprising 321 par- ticipants reported left ventricular ejection fraction at baseline and after ESA treatment, and the overall bene- ficial change was 6.88 (95% CI: 0.49–13.28, p = 0.03; Fig- ure 2 B). In three studies, administration of ESA therapy was correlated with a decrease in B-type natriuretic pro- tein (BNP) levels, with a mean change of –272.20 (95% CI:

(–444.52)–(–99.89), p = 0.002; Figure 2 C). The use of ESA therapy led to an improvement in NYHA functional class in five studies and the mean difference was –0.33 (95% CI: (–0.44)–(–0.23), p < 0.00001; Figure 2 D).

With regard to the type of exercise test, ESA therapy compared with control improved 6-minute walk dis- tance (6-MWD) by 81.48 m (95% CI: 14.57–148.39,

p = 0.02; Figure 2 E), exercise duration by 79.12 s

(95% CI: 14.53–143.72, p = 0.02; Figure 2 F), and peak

A. Comparison of hemoglobin

Study ESAs Control Weight Mean difference Mean difference

or subgroup Mean SD Total Mean SD Total (%) IV, random, 95% CI IV, random, 95% CI Ghali 2008 1.9 1 162 0.3 1 157 17.9 1.60 (1.38, 1.82)

Kourea 2008 1.6 1.3 21 –0.9 3.5 20 2.1 2.50 (0.87, 4.13) Mancini 2003 3.3 0.9 18 0.6 1.2 8 5.3 2.70 (1.77, 3.63) Palazzuoli 2006 1.2 0.5 20 –0.8 0.6 18 14.6 2.00 (1.65, 2.35) Palazzuoli 2007 2 0.7 26 –0.1 0.7 25 13.9 2.10 (1.72, 2.48) Parissis 2008 1.8 1.05 21 0.4 1 11 7.3 1.40 (0.66, 2.14) Ponikowski 2007 2.4 0.4 19 0.9 0.5 22 16.5 1.50 (1.22, 1.78) Silverberg 2001 2.6 1.2 16 –0.1 0.8 16 7.7 2.70 (1.99, 3.41) van Veldhuisen 2007 1.8 0.95 110 0 1.15 55 14.7 1.80 (1.45, 2.15)

Total (95% CI) 413 332 100.0 1.89 (1.64, 2.14)

Heterogeneity: t2 = 0.08; c2 = 22.69, df = 8 (p = 0.004); I2 = 65%

Test for overall effect: Z = 14.90 (p < 0.00001)

B. Comparison of LVEF

Study ESAs Control Weight Mean difference Mean difference

or subgroup Mean SD Total Mean SD Total (%) IV, random, 95% CI IV, random, 95% CI Kourea 2008 5 5 21 –5 12 20 18.9 10.00 (4.32, 15.68)

Palazzuoli 2007 6.8 6.75 26 –1.5 6.05 25 20.9 8.30 (4.78, 11.82) Parissis 2008 5 6 21 –3 5.5 11 20.4 8.00 (3.86, 12.14) Silverberg 2001 5.5 12.3 16 –5.4 7.3 16 17.6 10.90 (3.89, 17.91) van Veldhuisen 2007 –0.02 0.91 110 1.27 1.29 55 22.3 –1.29 (–1.67, –0.91)

Total (95% CI) 194 127 100.0 6.88 (0.49, 13.28)

Heterogeneity: t2 = 47.78; c2 = 72.75, df = 4 (p < 0.00001); I2 = 95%

Test for overall effect: Z = 2.11 (p =0.03)

C. Comparison of BNP

Study ESAs Control Weight Mean difference Mean difference

or subgroup Mean SD Total Mean SD Total (%) IV, random, 95% CI IV, random, 95% CI Kourea 2008 –340 433 21 179 685 20 15.5 –519.00 (–871.74, –166.26)

Palazzuoli 2006 –246 254 20 –47 294 18 30.5 –199.00 (–374.61, –23.39) Palazzuoli 2007 –246 253.5 26 –50 344 25 31.5 –196.00 (–362.37, –29.63) Parissis 2008 –441 1.029 21 424 705.5 11 6.8 –865.00 (–1471.23, –258.77) Ponikowski 2007 –90.5 269.4 19 –26.5 787.6 22 15.7 –64.00 (–414.70, 286.70)

Total (95% CI) 107 96 100.0 –272.20 (444.52, –99.89)

Heterogeneity: t2 = 17341.68; c2 = 7.92, df = 4 (p = 0.09); I2 = 50%

Test for overall effect: Z = 3.10 (p = 0.002)

Figure 2. Effects of erythropoiesis-stimulating agent therapy in heart failure patients with anemia at follow-up compared to baseline

–4 –2 0 2 4

Favours control Favours ESA

–50 –25 0 25 50

Favours control Favours ESA

–1000 –500 0 500 1000

Favours ESA Favours control

(4)

D. Comparison of NYHA functional class

Study ESAs Control Weight Mean difference Mean difference

or subgroup Mean SD Total Mean SD Total (%) IV, random, 95% CI IV, random, 95% CI Ghali 2008 –0.19 0.04 162 –0.13 0.04 157 34.7 –0.06 (–0.07, –0.05)

Palazzuoli 2006 –0.7 0.55 20 0.2 0.5 18 7.8 –0.90 (–1.23, –0.57) Palazzuoli 2007 –0.6 0.55 26 0.1 0.45 25 10.3 –0.70 (–0.98, –0.42) Parissis 2008 –0.7 0.45 21 0.5 0.55 11 6.4 –1.20 (–1.58, –0.82) Silverberg 2001 –0.6 0.55 16 0.4 0.5 16 6.8 –1.00 (–1.36, –0.64) van Veldhuisen 2007 –0.3 0.06 110 –0.23 0.08 55 34.1 –0.07 (–0.09, –0.05)

Total (95% CI) 355 282 100.0 –0.33 (–0.44, –0.23)

Heterogeneity: t2 = 0.01; c2 = 105.71, df = 5 (p < 0.00001); I2 = 95%

Test for overall effect: Z = 6.15 (p < 0.00001) –2 –1 0 1 2

Favours ESA Favours control

Figure 2. Cont.

E. Comparison of 6-minute hall walk

Study ESAs Control Weight Mean difference Mean difference

or subgroup Mean SD Total Mean SD Total (%) IV, random, 95% CI IV, random, 95% CI Kourea 2008 106 66 21 –40 79 20 22.0 146.00 (101.33, 190.67)

Mancini 2003 141 100 15 123 100 8 17.4 18.00 (–67.81, 103.81) Palazzuoli 2006 120 141 20 12 147 18 16.7 108.00 (16.18, 199.82) Parissis 2008 69 96 21 –47 91.5 11 19.5 116.00 (48.11, 183.89) van Veldhuisen 2007 34.2 7.3 110 11.4 10.3 55 24.4 22.80 (19.76, 25.84)

Total (95% CI) 187 112 100.0 81.48 (14.57, 148.39)

Heterogeneity: t2 = 4777.73; c2 = 39.48, df = 4 (p < 0.00001); I2 = 90%

Test for overall effect: Z = 2.39 (p = 0.02) –200 –100 0 100 200

Favours control Favours ESA

F. Comparison of exercise duration

Study ESAs Control Weight Mean difference Mean difference

or subgroup Mean SD Total Mean SD Total (%) IV, random, 95% CI IV, random, 95% CI Ghali 2008 57.3 29.3 162 46.5 30.6 157 29.9 10.80 (4.22, 17.38)

Mancini 2003 67 113 15 –83 143.5 8 15.4 150.00 (35.29, 264.71) Palazzuoli 2006 78 72 20 –19 57 18 26.7 97.00 (55.90, 138.10) Ponikowski 2007 38 50.5 19 –58 53 22 28.0 96.00 (64.28, 127.72)

Total (95% CI) 216 205 100.0 79.12 (14.53, 143.72)

Heterogeneity: t2 = 3622.91; c2 = 46.95, df = 3 (p < 0.00001); I2 = 94%

Test for overall effect: Z = 2.40 (p = 0.02) –200 –100 0 100 200

Favours control Favours ESA

G. Comparison of peak oxygen consumption

Study ESAs Control Weight Mean difference Mean difference

or subgroup Mean SD Total Mean SD Total (%) IV, random, 95% CI IV, random, 95% CI Mancini 2003 1.7 2.3 15 –0.5 1.75 8 30.0 2.20 (0.52, 3.88)

Palazzuoli 2006 2.6 2.8 20 –0.5 2.8 18 29.0 3.10 (1.32, 4.88) Ponikowski 2007 0 0.6 19 –0.5 0.55 22 40.9 0.50 (0.15, 0.85)

Total (95% CI) 54 48 100.0 1.77 (0.02, 3.51)

Heterogeneity: t2 = 1.90; c2 = 11.20, df = 2 (p = 0.004); I2 = 82%

Test for overall effect: Z = 1.98 (p = 0.05) –10 –5 0 5 10

Favours control Favours ESA

oxygen consumption (VO

2

) by 1.77  ml/kg/min (95% CI:

0.02–3.21, p = 0.05; Figure 2 G).

Hospitalizations and all-cause mortality

The hospitalization analysis demonstrated a  sig- nificant protective effect in the ESA treatment group compared with the control group (OR = 0.61, 95% CI:

0.39–0.94, p = 0.02; Figure 3 A), but there was no signif-

icant reduction in all-cause mortality (OR = 0.78, 95% CI:

0.51–1.21, p = 0.27; Figure 3 B).

Potential sources of heterogeneity analysis

A random-effect univariate meta-regression analysis

for the hemoglobin level change, hospitalizations and all-

cause mortality in heart failure was conducted to explore

the potential sources of heterogeneity. Data on the age,

(5)

A. Heart failure hospitalizations

Study ESAs Control Weight Odds ratio Odds ratio or subgroup Events Total Events Total (%) IV, random, 95% CI IV, random, 95% CI

Cleland 2005 3 18 0 6 1.9 2.94 (0.13, 65.26)

Ghali 2008 25 162 31 157 21.6 0.74 (0.42, 1.32)

Kourea 2008 3 21 5 20 6.2 0.50 (0.10, 2.44)

Mancini 2003 1 16 4 8 2.9 0.07 (0.01, 0.78)

Palazzuoli 2006 4 20 8 18 7.3 0.31 (0.07, 1.31)

Palazzuoli 2007 4 26 8 25 7.9 0.39 (0.10, 1.50)

Parissis 2008 2 21 3 11 4.3 0.28 (0.04, 2.01)

Ponikowski 2007 4 19 9 22 7.6 0.39 (0.10, 1.55)

Swedberg 2013 314 1136 311 1142 33.1 1.02 (0.85, 1.23) van Veldhuisen 2007 4 110 4 55 7.3 0.48 (0.12, 2.00)

Total (95% CI) 1549 1464 100.0 0.61 (0.39, 0.94)

Total events 364 383

Heterogeneity: t2 = 0.14, c2 = 14.68, df = 9 (p = 0.10), I2 = 39%

Test for overall effect: Z = 2.25 (p = 0.02)

B. All-cause mortality

Study ESAs Control Weight Odds ratio Odds ratio or subgroup Events Total Events Total (%) IV, random, 95% CI IV, random, 95% CI

Cleland 2005 1 18 1 6 2.1 0.29 (0.02, 5.60)

Ghali 2008 11 162 18 157 20.6 0.56 (0.26, 1.23)

Kourea 2008 1 21 3 20 3.3 0.28 (0.03, 2.98)

Mancini 2003 1 15 0 8 1.7 1.76 (0.06, 48.19)

Palazzuoli 2006 1 20 2 18 2.9 0.42 (0.03, 5.08)

Palazzuoli 2007 2 26 3 25 5.0 0.61 (0.09, 4.01)

Parissis 2008 0 21 2 11 1.9 0.09 (0.00, 2.02)

Ponikowski 2007 1 19 1 22 2.3 1.17 (0.07, 20.02)

Silverberg 2001 0 16 4 16 2.0 0.08 (0.00, 1.71)

Swedberg 2013 388 1136 376 1142 56.0 1.06 (0.89, 1.26) van Veldhuisen 2007 6 108 0 55 2.2 7.04 (0.39, 127.28)

Total (95% CI) 1562 1480 100.0 0.78 (0.51, 1.21)

Total events 412 410

Heterogeneity: t2 = 0.08, c2 = 11.71, df = 10 (p = 0.30), I2 = 15%

Test for overall effect: Z = 1.10 (p = 0.27)

Figure 3. Effect of erythropoiesis-stimulating agent therapy on hospitalizations and mortality

0.05 0.2 1 5 20

Favours ESA Favours control

0.05 0.2 1 5 20

Favours ESA Favours control

sex (% male), baseline EF, baseline hemoglobin and ESA therapy (DA or EPO) were included. As a result, baseline hemoglobin was the major heterogeneity source iden- tified for hemoglobin level change (adjusted R

2

= 0.21,

p = 0.089). The ESA therapy was the major heterogeneity

source identified for hospitalizations (adjusted R

2

= 0.28,

p = 0.093) and all-cause mortality (adjusted R2

= 0.23,

p = 0.035). Age, sex and baseline EF may not contrib-

ute to the source of heterogeneity for hemoglobin level change, hospitalizations and all-cause mortality (p > 0.1).

Discussion

Anemia in HF is related to adverse clinical outcomes, but little is known about the effects of its treatment with ESA on cardiac dimensions and function. Heart failure is associated with elevated pro-inflammatory cytokines, which cause not only decreased erythropoietin (EPO) production, but also resistance to its actions on bone marrow [26, 27]. Erythropoietin levels in HF patients are

lower than expected, which is possibly attributed to the action of pro-inflammatory cytokines [26, 28]. Chronic kidney disease or milder forms of renal dysfunction are also common in HF patients, and may contribute to de- creased EPO production. Other factors such as inflam- mation, diabetes, hemo-dilution, gastrointestinal mal- absorption and blood loss, absolute and functional iron deficiency, and drugs such as angiotensin receptor block- ers (ARB) and angiotensin-converting enzyme inhibitors (ACEI) are believed to contribute to the development of anemia in this patient population [29]. Previous studies have shown that ESA therapy can improve heart function, exercise capacity, and quality of life in HF patients with anemia. However, because of some deficiencies such as small sample sizes and lack of double-blind and/or pla- cebo-controlled design in those studies, no very definite conclusions could be drawn.

In this meta-analysis of 11 RCTs with 3044 patients,

we found that ESA therapy leads to a significant improve-

ment in LVEF and BNP compared with placebo. Also we

(6)

found that ESA therapy reduced the NYHA functional class, an effect that was partly associated with patient symptomatic improvement. The specific mechanism of the improvement is not very clear. Previous studies have shown that ESA therapy seems to have potential effects to enhance cardiac contractile function and improve car- diac remodeling through its angiogenic and anti-apoptot- ic properties [30, 31].

Our meta-analysis found that the ESA therapy ap- proach leads to a significant improvement in exercise ca- pacity assessed by 6-MWD, exercise duration, and peak VO

2

. There are a number of potential mechanisms such as the treatment of anemia, attenuation of peripheral hy- poxia with the concomitant improvement of metabolic status of peripheral muscles and the reduction of volume overloading which may explain the beneficial effects of ESA on exercise tolerance and quality of life in anemic HF patients [17].

As compared with placebo, ESA therapy led to a sig- nificant increase in hemoglobin levels and reduction in hospitalizations. However, we found that ESA therapy was not associated with a  significant reduction in all- cause mortality compared with placebo. This observa- tion is partly in agreement with the previous studies [32], which suggest that ESA therapy can improve the symptoms of heart failure but does little to reduce all- cause mortality.

Despite employing a random effects model, statis- tical heterogeneity in the present study was consider- able. There are many factors contributing to heteroge- neity, such as the baseline disease severity, eligibility criteria, the patient selection and randomization, dif- ferences in trial designs, the inclusion of different in- terventions, the follow-up in the trials and differences in sample size. All of these factors lead to increase in heterogeneity.

Conclusions

We found that the treatment of anemia with ESA therapy did not reduce the rate of all-cause mortali- ty among patients with heart failure. However, a trend of lower BNP, higher LVEF, reduction in hospitaliza- tions and improvement in NYHA functional class was observed. At the same time, ESA therapy led to an in- crease of exercise tolerance. Consequently, our findings suggest that treatment of anemia with ESA improved the symptoms of heart failure but did not improve clin- ical outcomes.

Acknowledgments

Hengliang Zhang and Pei Zhang contributed equally to this work.

Conflict of interest

The authors declare no conflict of interest.

References

1. Lindenfeld J. Prevalence of anemia and effects on mortality in patients with heart failure. Am Heart J 2005; 149: 391-401.

2. Ghali JK. Anemia and heart failure. Curr Opin Cardiol 2009; 24:

172-8.

3. Caira C, Ansalone G, Mancone M, et al. Heart failure and iron de- ficiency anemia in Italy: results from CARMES-1 registry. Future Cardiol 2013; 9: 437-44.

4. Beutler E, Waalen J. The definition of anemia: what is the lower limit of normal of the blood hemoglobin concentration? Blood 2006; 107: 1747-50.

5. Groenveld HF, Januzzi JL, Damman K, et al. Anemia and mortality in heart failure patients a systematic review and meta-analysis.

J Am Coll Cardiol 2008; 52: 818-27.

6. Salisbury AC, Kosiborod M. Outcomes associated with anemia in patients with heart failure. Heart Fail Clin 2010; 6: 359-72.

7. Horwich TB, Fonarow GC, Hamilton MA, et al. Anemia is asso- ciated with worse symptoms, greater impairment in functional capacity and a significant increase in mortality in patients with advanced heart failure. J Am Coll Cardiol 2002; 39: 1780-6.

8. Kalra PR, Bolger AP, Francis DP, et al. Effect of anemia on exercise tolerance in chronic heart failure in men. Am J Cardiol 2003; 91:

888-91.

9. Zuccala G, Marzetti E, Cesari M, et al. Correlates of cognitive im- pairment among patients with heart failure: results of a multi- center survey. Am J Med 2005; 118: 496-502.

10. Adams KF Jr, Pina IL, Ghali JK, et al. Prospective evaluation of the association between hemoglobin concentration and quality of life in patients with heart failure. Am Heart J 2009; 158: 965-71.

11. Unger EF, Thompson AM, Blank MJ, et al. Erythropoiesis-stimu- lating agents: time for a reevaluation. N Engl J Med 2010; 362:

189-92.

12. Phrommintikul A, Haas SJ, Elsik M, et al. Mortality and target haemoglobin concentrations in anaemic patients with chronic kidney disease treated with erythropoietin: a  meta-analysis.

Lancet 2007; 369: 381-8.

13. Kimel M, Leidy NK, Mannix S, et al. Does epoetin alfa improve health-related quality of life in chronically ill patients with ane- mia? Summary of trials of cancer, HIV/AIDS, and chronic kidney disease. Value Health 2008; 11: 57-75.

14. van der Meer P, Lok DJ, Januzzi JL, et al. Adequacy of endogenous erythropoietin levels and mortality in anaemic heart failure pa- tients. Eur Heart J 2008; 29: 1510-5.

15. Cleland JG, Sullivan JT, Ball S, et al. Once-monthly administration of darbepoetin alfa for the treatment of patients with chronic heart failure and anemia: a pharmacokinetic and pharmacody- namic investigation. J Cardiovasc Pharmacol 2005; 46: 155-61.

16. Ghali JK, Anand IS, Abraham WT, et al. Randomized double-blind trial of darbepoetin alfa in patients with symptomatic heart fail- ure and anemia. Circulation 2008; 117: 526-35.

17. Kourea K, Parissis JT, Farmakis D, et al. Effects of darbepoetin-al- pha on quality of life and emotional stress in anemic patients with chronic heart failure. Eur J Cardiovasc Prev Rehabil 2008;

15: 365-9.

18. Mancini DM, Katz SD, Lang CC, et al. Effect of erythropoietin on exercise capacity in patients with moderate to severe chronic heart failure. Circulation 2003; 107: 294-9.

19. Palazzuoli A, Silverberg D, Iovine F, et al. Erythropoietin improves anemia exercise tolerance and renal function and reduces B-type natriuretic peptide and hospitalization in patients with heart failure and anemia. Am Heart J 2006; 152: 1096 e9-15.

(7)

20. Palazzuoli A, Silverberg DS, Iovine F, et al. Effects of beta-eryth- ropoietin treatment on left ventricular remodeling, systolic func- tion, and B-type natriuretic peptide levels in patients with the cardiorenal anemia syndrome. Am Heart J 2007; 154: 645 e9-15.

21. Parissis JT, Kourea K, Panou F, et al. Effects of darbepoetin alpha on right and left ventricular systolic and diastolic function in anemic patients with chronic heart failure secondary to isch- emic or idiopathic dilated cardiomyopathy. Am Heart J 2008;

155: 751 e1-7.

22. Ponikowski P, Anker SD, Szachniewicz J, et al. Effect of darbep- oetin alfa on exercise tolerance in anemic patients with symp- tomatic chronic heart failure: a randomized, double-blind, place- bo-controlled trial. J Am Coll Cardiol 2007; 49: 753-62.

23. Silverberg DS, Wexler D, Sheps D, et al. The effect of correction of mild anemia in severe, resistant congestive heart failure using subcutaneous erythropoietin and intravenous iron: a  random- ized controlled study. J Am Coll Cardiol 2001; 37: 1775-80.

24. Swedberg K, Young JB, Anand IS, et al. Treatment of anemia with darbepoetin alfa in systolic heart failure. N Engl J Med 2013;

368: 1210-9.

25. van Veldhuisen DJ, Dickstein K, Cohen-Solal A, et al. Random- ized, double-blind, placebo-controlled study to evaluate the ef- fect of two dosing regimens of darbepoetin alfa in patients with heart failure and anaemia. Eur Heart J 2007; 28: 2208-16.

26. Opasich C, Cazzola M, Scelsi L, et al. Blunted erythropoietin pro- duction and defective iron supply for erythropoiesis as major causes of anaemia in patients with chronic heart failure. Eur Heart J 2005; 26: 2232-7.

27. Belonje AM, Voors AA, van Gilst WH, et al. Erythropoietin in chronic heart failure. Congest Heart Fail 2007; 13: 289-92.

28. Belonje AM, Voors AA, van der Meer P, et al. Endogenous eryth- ropoietin and outcome in heart failure. Circulation 2010; 121:

245-51.

29. Arora NP, Ghali JK. Anemia and iron deficiency in heart failure.

Heart Fail Clin 2014; 10: 281-94.

30. van der Meer P, Lipsic E, Henning RH, et al. Erythropoietin in- duces neovascularization and improves cardiac function in rats with heart failure after myocardial infarction. J Am Coll Cardiol 2005; 46: 125-33.

31. Calvillo L, Latini R, Kajstura J, et al. Recombinant human eryth- ropoietin protects the myocardium from ischemia-reperfusion injury and promotes beneficial remodeling. Proc Natl Acad Sci USA 2003; 100: 4802-6.

32. Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med 2009; 361: 2019-32.

Cytaty

Powiązane dokumenty

Prospective random- ized multicenter study comparing cyclosporin alone versus the combination of antithymocyte globulin and cyclosporin for treat- ment patients with

The aim of the study was to evaluate the effect of L-citrulline supplementation on functional class, ejection fraction and peripheral blood flow in patients with systolic

The aim of our study was to investigate the use of L without a load- ing dose in a cohort of end-stage CHF patients ad- mitted to our institution for acute decompensation HF compared

Our study demonstrated that there is a statistically significant correlation between homocysteine plasma levels and the severity of heart failure in patients with prior

two-minutes dark adaptation (R1); latency for the onset of constriction (T1); minimum pupil radius af- ter pupil’s reaction to light (R2); maximum constric- tion velocity

Conclusions: In patients with narrow QRS complex and baseline mechanical asynchrony, who underwent CRT after optimal medical management, there was a significant reduction in NYHA

Predis- chargwe initiation of carvedilol in patients hospitalized for decompensated heart failure: Results of the Initi- ation Management Predischarge: process for Assess- ment

This study aimed to evaluate the effect of endurance training and a combination of en- durance and resistance training (combined exercise), functional electrical stimulation (FES),