Abstract
Background and Aims
Evidence is lacking that correcting iron deficiency (ID) has clinically important benefits for patients with heart failure with preserved ejection fraction (HFpEF).
Methods
FAIR-HFpEF was a multicentre, randomized, double-blind trial designed to compare intravenous ferric carboxymaltose (FCM) with placebo (saline) in 200 patients with symptomatic HFpEF and ID (serum ferritin < 100 ng/mL or ferritin 100–299 ng/mL with transferrin saturation < 20%). The primary endpoint was change in 6-min walking test distance (6MWTD) from baseline to week 24. Secondary endpoints included changes in New York Heart Association class, patient global assessment, and health-related quality of life (QoL).
Results
The trial was stopped because of slow recruitment after 39 patients had been included (median age 80 years, 62% women). The change in 6MWTD from baseline to week 24 was greater for those assigned to FCM compared to placebo [least square mean difference 49 m, 95% confidence interval (CI) 5–93; P = .029]. Changes in secondary endpoints were not significantly different between groups. The total number of adverse events (76 vs. 114) and serious adverse events (5 vs. 19; rate ratio 0.27, 95% CI 0.07–0.96; P = .043) was lower with FCM than placebo.
Conclusions
In patients with HFpEF and markers of ID, intravenous FCM improved 6MWTD and was associated with fewer serious adverse events. However, the trial lacked sufficient power to identify or refute effects on symptoms or QoL. The potential benefits of intravenous iron in HFpEF with ID should be investigated further in a larger cohort.
Keywords: Heart failure, Preserved ejection fraction, Exercise capacity, Iron deficiency
Structured Graphical Abstract
See the editorial comment for this article ‘Intravenous iron therapy in heart failure with preserved ejection fraction: how far have we walked?’, by V. Kittipibul and R.J. Mentz, https://doi.org/10.1093/eurheartj/ehae490.
Introduction
In chronic heart failure (HF), iron deficiency (ID) is prevalent in up to 50% of all patients.1–3 As a very frequent comorbidity in HF, ID has garnered significant research attention in recent years. This has led to the conduct of several large-scale trials,4–6 reinforcing that correcting ID improves quality of life (QoL), enhances exercise capacity, and reduces HF hospitalization rates in patients with HF with reduced ejection fraction (HFrEF) or mildly reduced ejection fraction, leading to guideline recommendations for the use of ferric carboxymaltose (FCM) and ferric derisomaltose exclusively in these populations.7,8
A treatment gap persists for patients with HF with preserved ejection fraction (HFpEF), and it has thus not been demonstrated whether recommended diagnostic criteria apply in this cohort of patients. This point is important, because besides the use of diuretics and the recently introduced successful treatment with sodium–glucose co-transporter 2 (SGLT2) inhibitors,9 the treatment of comorbidities such as coronary artery disease, atrial fibrillation (AF), arterial hypertension, chronic obstructive pulmonary disease, or obesity remains the mainstay of therapy in HFpEF.10,11 With guideline-recommended threshold values for detecting ID in patients with HF set at serum ferritin < 100 ng/mL or serum ferritin 100–299 ng/mL with transferrin saturation (TSAT) < 20%, recent meta-analysis data from 15 studies suggest that the prevalence of ID in patients with HFpEF reaches 59%.12 We designed the multicentre, randomized, double-blind FAIR-HFpEF trial to evaluate the effectiveness of FCM in patients with HFpEF and ID on exercise capacity as assessed by the 6-min walking test distance (6MWTD).
Methods
FAIR-HFpEF was a prospective, multicentre, 1:1 randomized, double-blind, placebo-controlled trial investigating the effects of intravenous FCM on exercise tolerance, symptoms, and QoL in patients with HFpEF and ID (defined as ferritin < 100 ng/mL or ferritin 100–299 ng/mL plus TSAT < 20%), with or without anaemia. A detailed trial protocol has been previously published.13
In brief, treatment was provided after randomization at baseline in 1–2 sessions (1000–2000 mg FCM or saline) as well as at week 16 (500–1000 mg) and at week 32 (500–1000 mg), always in a setting that kept the blinding (see also Supplementary data online, Table S1). Treatment according to randomization was continued at weeks 16 and 32 unless ferritin was >800 ng/mL, or when ferritin was > 500 ng/mL with TSAT > 50%, or when haemoglobin was > 16.0 g/dL at any stage during follow-up (in which case saline had to be given). These stopping rules were identical to that of FAIR-HF and almost as liberal as in FAIR-HF2.14
Randomization in FAIR-HFpEF included stratification by presence of AF at baseline and by screening haemoglobin. The trial enrolled men and women aged ≥18 years with chronic HFpEF and diminished exercise capacity (defined as a 6MWTD < 450 m, averaged from the last two documented tests within 8 weeks prior to planned randomization), New York Heart Association (NYHA) class II–III symptoms, treated with a diuretic, elevated natriuretic peptide levels or a history of HF-related hospitalization within 12 months prior to randomization, and a left ventricular ejection fraction (LVEF) ≥ 45%. All procedures were conducted in accordance with the principles of the Declaration of Helsinki and International Conference on Harmonization Good Clinical Practice. Funding for the trial was provided by Vifor Pharma, Switzerland. Charité—Universitätsmedizin Berlin served as the sponsor of the trial. An independent ethics committee approved the protocol at all participating centres, and all subjects provided written informed consent. The trial was registered at ClinicalTrials.gov (NCT03074591).
The original protocol aimed to randomize a total of 200 patients, evenly split between those with and without anaemia. Of note, the number of patients randomized in the 2.25 years before COVID started in March 2020 was 19, and in the following 1.5 years, we randomized 20 patients. This means there was no negative impact of COVID on recruitment. Budget constraints and regulatory problems and most likely also several competing trials in HFpEF prevented the expansion of enrolment to sites beyond Germany (Supplementary data online, Appendix S1) and caused overall very slow patient recruitment. As a result, enrolment was halted after recruiting 74 patients, of whom 40 were randomized (Figure 1).
Trial procedures
With FCM being a brown substance and normal saline serving a comparator, the trial was conducted by unblinded and blinded investigators. Infusion sites and treatment vials were covered, and if considered needed, also eye masks were used during infusions to ensure patient blinding. Following screening (visit 1), study-related assessments, including the 6MWTD, patient global assessment (PGA), QoL questionnaires, and NYHA class, were performed by blinded investigators at baseline, after 8 ± 3 days, and after 4, 16 ± 1, 24 ± 1, 32 ± 2, and 52 ± 2 weeks (visits 2–7).13 Health-related quality of life (HRQoL) was evaluated using the HRQoL questionnaire, which integrates the European QoL-5 Dimensions (EQ-5D) questionnaire as a generic instrument with a disease-specific cardiology tool, the Kansas City Cardiomyopathy Questionnaire (KCCQ). Unblinded investigators performed infusions of intravenous placebo (i.e. normal saline) or FCM according to the dosing regimen in Supplementary data online, Table S1 as well as certain trial-related procedures (e.g. drug accountability, monitoring patients for elevated iron parameters or haemoglobin levels).
Primary and secondary endpoints
The primary endpoint of the trial was the change in 6MWTD from baseline to week 24, assessed in metres. Secondary endpoints included the change in 6MWTD from baseline to visits 3–7 and the change in PGA, NYHA class, blood parameters of kidney function and inflammation, and HRQoL from baseline to visits 3–7, and as safety-related outcome the rate of recurrent cardiovascular hospitalizations.13
Statistical analysis
A sample size of 86 patients per group gave a power of 90% for a twosample ttest at the usual one-sided level of 2.5% if the standardized mean difference (Cohen's d) is 0.50. Accounting for 10% dropout the original aim was to recruit 100 patients per group. Enrolment, however, proved to be more difficult than expected for reasons outlined above.
The primary analysis was based on the modified intention-to-treat (ITT) population including all randomized patients who received at least one dose of study medication and had at least one post-baseline assessment of the primary endpoint; they were analysed in the group they were randomized to. The primary endpoint was analysed using a mixed-model repeated measures (MMRM) approach adjusted for the presence of AF, anaemia, age at baseline, visit, and baseline 6MWTD; visit-by-treatment interactions were also included. Least square group differences were reported with standard error, 95% confidence intervals (CIs), and P-values testing the null hypothesis of no intervention effect using all available data without any imputation method. The analyses of continuous secondary endpoints followed the same lines as the analysis of the primary endpoint. Patient global assessment at all visits was compared between both groups using Fisher's exact tests. To adjust for covariates, a mixed-effects proportional odds model including a random intercept for subject and the main effects for treatment, visit, treatment–visit interaction, presence of AF at baseline, haemoglobin status at baseline, and age at baseline, as fixed effects was calculated. The change in NYHA class from baseline to the respective visit in the FCM group was tested using the Bowker symmetry test. Between groups, the NYHA class was compared using the Wilcoxon–Mann–Whitney test. As with PGA, mixed-effects proportional odds models were used for adjusted analyses. Recurrent events were modelled by negative binomial regression. Adverse events were summarized as frequencies and percentages by treatment group. Event rates were compared using negative binomial regression accounting for over-dispersion. All analyses were carried out using the statistical software R (version 4.3.1).
Results
A total of 74 patients with HFpEF, reduced exercise capacity, and ID were screened. Of them, 42 patients were eligible for randomization in six German centres, three of whom were excluded due to presence of exclusion criteria discovered after electronic randomization in two patients (i.e. they were considered randomized in error) and for personal reasons in one patient immediately after the first treatment application (with no endpoint data available after baseline). The final analysis was therefore performed in 39 patients in a modified ITT analysis per statistical analysis plan, 18 of whom received FCM and 21 received placebo/saline (Figure 1).
For the overall cohort, the median age was 80 years [inter-quartile range (IQR): 75–84], 62% were women, haemoglobin was 12.5 g/dL (IQR: 11.4–13.1), serum ferritin was 49.0 ng/mL (IQR: 24.7–91.0), TSAT was 17.0% (IQR: 13.7–22.6), and median N-terminal pro-B-type natriuretic peptide was 772 pg/mL (IQR: 342–1499). Seven of 39 patients had an LVEF of <50%. The two treatment groups were similar with regard to age, sex, haemoglobin, serum ferritin, TSAT, other laboratory characteristics, and use of cardiovascular medications and 6MWTD at baseline (all P > .05). Detailed clinical characteristics are presented in Table 1.
Table 1.
Placebo/saline (n = 21) |
FCM (n = 18) |
|
---|---|---|
Sex (women) | 14 (67%) | 10 (56%) |
Weight (kg) | 83.0 (75.0–90.0) | 83.0 (75.8–87.8) |
BMI (kg/m2) | 29.8 (26.1–32.5) | 28.9 (26.3–32.4) |
BMI ≥ 30 kg/m2 | 10 (48%) | 6 (33%) |
Age (years) | ||
Mean (SD) | 79 ± 7.03 | 76 ± 8.88 |
Median (Q1–Q3) | 80 (77–84) | 79 (72–83) |
LVEF (%) | ||
Mean (SD) | 55.1 ± 7.8 | 55.3 ± 6.5 |
Median (Q1–Q3) | 55 (50–56) | 55 (50–60) |
NYHA class | ||
II | 10 (48%) | 11 (61%) |
III | 11 (52%) | 7 (39%) |
Cause of heart failure | ||
Ischaemic | 14 (67%) | 9 (50%) |
Non-ischaemic | 7 (33%) | 9 (50%) |
Haemodynamics | ||
Heart rate (b.p.m.) | 68 (61–80) | 68 (60–71) |
Systolic blood pressure (mmHg) | 125 (111–136) | 127 (113–151) |
Diastolic blood pressure (mmHg) | 67 (62–72) | 75 (63–80) |
Laboratory results | ||
Haemoglobin (g/dL) | 12.0 (11.4–12.8) | 12.9 (11.7–13.2) |
Serum ferritin (ng/mL) | 50 (25–94) | 44 (23–72) |
Serum ferritin < 100 ng/mL | 17 (81%) | 15 (83%) |
TSAT (%) | 16.0 (14.0–21.0) | 19.9 (13.8–24.8) |
TSAT < 20% | 14 (67%) | 9 (50%) |
Creatinine (mg/dL) | 1.1 (0.8–1.5) | 1.3 (1.0–1.6) |
eGFR (mL/min/1.73 m2) | 59 (36–76) | 40 (32–66) (n = 17) |
Bilirubin (mg/dL) | 0.7 (0.5–0.9) (n = 10) | 0.5 (0.4–0.6)(n = 7) |
Blood urea nitrogen (mg/dL) | 10 (8–18) (n = 9) | 19 (10–33) (n = 6) |
ASAT (U/L) | 24 (22–27) | 25 (22–28) |
ALAT (U/L) | 19 (15–24) | 16 (14–22) |
γ-GT (U/L) | 27 (17–46) | 31 (19–53) (n = 17) |
C-reactive protein (mg/L) | 2.9 (1.4–6.0) | 2.1 (1.8–5.0) |
NT-proBNP (pg/mL) | 490 (343–1331) | 1129 (407–2293) |
Medical history | ||
Previous hospitalization for HF | 5 (24%) | 9 (50%) |
Atrial fibrillation/flutter | 10 (48%) | 10 (56%) |
Diabetes mellitus | 8 (38%) | 10 (56%) |
Hypertension | 19 (90%) | 16 (89%) |
Dyslipidaemia | 14 (67%) | 10 (56%) |
Coronary artery disease | 14 (67%) | 9 (50%) |
Previous myocardial infarction | 7 (33%) | 1 (6%) |
Previous CABG | 2 (10%) | 2 (11%) |
Previous percutaneous coronary intervention | 12 (57%) | 6 (33%) |
Valvular heart disease (deemed clinically meaningful) | 12 (57%) | 11 (61%) |
Treatment | ||
ACE inhibitor | 13 (62%) | 6 (33%) |
ARB | 6 (29%) | 5 (28%) |
ARNi | 0 (0%) | 2 (11%) |
Beta-blocker | 16 (76%) | 13 (72%) |
Calcium antagonist | 7 (33%) | 3 (17%) |
MRA | 7 (33%) | 2 (11%) |
SGLT2 inhibitor | 4 (19%) | 1 (6%) |
Any other anti-diabetic | 7 (33%) | 6 (33%) |
Loop diuretic | 14 (67%) | 15 (83%) |
Insulin | 3 (14%) | 3 (17%) |
Any other diuretic | 6 (29%) | 4 (22%) |
Allopurinol | 5 (24%) | 8 (44%) |
Proton-pump inhibitor | 11 (52%) | 7 (39%) |
Cholesterol-lowering drug | 16 (76%) | 10 (56%) |
Any anti-platelet | 10 (48%) | 7 (39%) |
Any anticoagulant | 10 (48%) | 9 (50%) |
Previous ICD | 1 (5%) | 1 (6%) |
Previous cardiac resynchronization therapy | 3 (14%) | 0 (0%) |
ALAT, alanine aminotransferase; ARB, angiotensin receptor blocker; ARNi, angiotensin receptor-neprilysin inhibitor; ASAT, aspartate aminotransferase; CABG, coronary artery bypass graft; eGFR, estimated glomerular filtration rate; ICD, implantable cardioverter defibrillator; MRA, mineralocorticoid receptor antagonist; γ-GT, γ-glutamyltransferase.
Follow-up
Of the 18 patients assigned to receive FCM, one did not complete the 24 weeks and the 52 weeks of follow-up. Of the 21 patients assigned to receive placebo/saline, 3 did not complete the 24 weeks of follow-up and 4 did not complete the 52 weeks of follow-up.
Primary and secondary endpoints
The 6MWTD improved in the FCM group from a median of 308 m (IQR: 198–378) at baseline to 403 m (IQR: 306–416) at week 24, with a change of 45 ± 62 m compared to baseline (P = .014) The respective values in the placebo/saline group were 325 m (IQR: 250–342) at baseline and 308 m (IQR: 285–354) at week 24, with a change of −8 ± 61 m vs. baseline (P = .62). The difference in least square means between the two groups at 24 weeks was 49 ± 22 m (mean ± SEM; 95% CI 5–93, P = .029). Of note, in total, 31 patients performed 6MWTD at week 24 (FCM: 15, placebo 16; see Supplementary data online, Table S2), but all 39 randomized patients contributed data to the result in the MMRM analysis for this endpoint. Detailed data are provided in Figure 2A and Table 2, which also show that the efficacy was somewhat further enhanced at week 32 (treatment effect between groups: 65 ± 22 m, P = .005) and then mostly lost at 52 weeks (treatment effect 13 ± 23 m, P = .57). Improvements in 6MWTD were similar in patients with and without anaemia (i.e. haemoglobin values of < 12.0 or ≥ 12.0 g/dL) at baseline (Figure 2B). When key subgroups were considered (including sex, ischaemic aetiology of HF, NYHA class, glomerular filtration rate, and haematinics at baseline), no significant interaction was found for subgroup of patients with HFpEF to respond differently to FCM vs. placebo with regard to changes in 6MWTD between baseline and 24 weeks (all P > .3; Supplementary data online, Figure S1); however, all subgroups were small. Due to these small subgroup sizes, the absence of interaction should not be considered as evidence of no interaction.
Table 2.
Baseline | Week 24 | Treatment effect with 95% CI (or OR) (at week 24) |
P-value | |||
---|---|---|---|---|---|---|
Placebo (n = 21) | FCM (n = 18) | Placebo (n = 18) | FCM (n = 17) | |||
Primary endpoint | ||||||
6 min walk test distance (m) | 325 (250–342) | 308 (198–378) | 308 (285–354) Change from BL to w24: −7.8 ± 60.6 | 403 (306–416) Change from BL to w24: 44.9 ± 61.9 | 49 (5.1, 93.0) | .029 |
Secondary endpoints | ||||||
Patient global assessment | - | - | Much worse: 1 Worse: 1 Little worse: 2 Unchanged: 6 Little improved: 5 Improved: 3 Much improved: 0 |
Much worse: 0 Worse: 1 Little worse: 2 Unchanged: 9 Little improved: 0 Improved: 4 Much improved: 1 |
OR 0.924 (0.24, 3.51) | .91 |
NYHA class | 2.52 ± 0.51 II: 10 III: 11 |
2.38 ± 0.50 II: 11 III: 7 |
2.50 ± 0.51 II: 9 III: 9 |
2.35 ± 0.49 II: 11 III: 6 |
−0.03 (−0.32, 0.26) | .83 |
EQ-5D-3L | 0.68 (0.59–0.76) | 0.62 (0.50–0.71) | 0.69 (0.60–0.78) | 0.68 (0.50–0.78) | −0.03 (−0.14, 0.09) | 0.63 |
KCCQ overall summary score | 72 (50–77) | 50 (36–66) | 70 (54–82) Change from BL to w24: 0.49 ± 14.81 | 60 (56–81) Change from BL to w24: 12.41 ± 11.53 | 6.5 (−3.8, 16.7) | .21 |
eGFR (mL/min/1.73 m2) | 59 (36–76) | 40 (32–66) | 50 (35–72) | 47 (38–63) | 0.27 (−7.16, 7.70) | .94 |
C-reactive protein (mg/L) | 2.9 (1.3–6.7) | 2.1 (1.8–6.1) | 2.5 (0.8–5.0) | 2.3 (1.5–8.3) | −8.2 (−32.6, 16.2) | .50 |
Other endpoints and biomarker assessments | ||||||
Haemoglobin (g/dL) | 12.0 (11.4–12.8) | 12.9 (11.7–13.2) | 12.1 (11.2–13.1) | 13.8 (13.4–14.2) | 0.93 (0.10, 1.76) | .028 |
Ferritin (µg/L) | 50 (25–94) | 44 (23–72) | 46 (29–78) | 326 (238–402) | 215 (141, 289) | <.001 |
TSAT (%) | 16.0 (14.0–21.0) | 19.9 (13.8–24.8) | 19.0 (13.5–23.0) | 30.0 (24.5–35.6) | 9.7 (3.9, 15.5) | .001 |
Systolic blood pressure (mmHg) | 125 (111–136) | 127 (113–151) | 130 (113–135) | 127 (118–139) | 1 (−10.3, 12.6) | .84 |
Diastolic blood pressure (mmHg) | 67 (62 –72) | 75 (63–81) | 71 (64–77) | 71 (63–79) | −4 (−10.3, 3.0) | .28 |
Heart rate (b.p.m.) | 68 (61–80) | 68 (60–71) | 74 (64–82) | 66 (60–67) | −4 (−12, 33) | .26 |
ASAT (U/L) | 24 (22–27) | 25 (22–28) (n = 17) |
24 (21–31) | 26 (23–27) (n = 16) |
−0.2 (−8.3, 8.7) | .96 |
ALAT (U/L) | 19 (15–24) | 16 (14–22) (n = 17) |
20 (17–25) | 22 (17–28) (n = 16) |
2.4 (−3.0, 7.8) | .38 |
γ−GT (U/L) | 27 (17–46) | 31 (19–53) (n = 17) |
30 (16–52) | 54 (33–105) (n = 16) |
21.4 (−5.4, 48.3) | .12 |
Creatinine (mg/dL) | 1.1 (0.8–1.5) | 1.3 (1.0–1.6) | 1.1 (0.9–1.5) | 1.2 (1.1–1.6) | −0.09 (−0.23, 0.16) | .53 |
Blood urea nitrogen (mg/dL) | 9.8 (7.5–18.2) (n = 9) |
18.9 (9.7–32.8) (n = 6) |
11.8 (9.7–15.6) (n = 8) |
19.2 (10.3–45.7) (n = 9) |
0.1 (−10.8, 11.1) | .98 |
BL, baseline; OR, odds ratio.
By week 24, 5 of 17 patients in the group receiving FCM reported moderate or much improvement in the PGA (in the placebo/saline group 3 of 18) with no significant difference compared to the group that received placebo/saline where 8 of 18 patients reported little or moderate improvement (in the FCM group 5 of 17) (P = .20) (see Supplementary data online, Figure S2).
At baseline, 11 (61%) of 18 patients treated with FCM were in NYHA class II and 7 (39%) in NYHA class III. At week 24, 11 (65%) of 17 patients were in NYHA class II and 6 (35%) in NYHA class III. For patients receiving placebo/saline, the respective numbers at baseline were 10 (48%) of 21 patients in NYHA class II and 11 (52%) in NYHA class III. Like in the FCM group, values were essentially unchanged in week 24 with 9 (50%) of 18 patients in NYHA class II and 9 (50%) patients in NYHA class III.
Between baseline and week 24 or other time points, no significant difference was noted between FCM and placebo/saline with regard to changes in the values of the EQ-5D or KCCQ (Figure 3). We observed a non-significant difference in the KCCQ overall summary score of 6.5 ± 5.1 points between FCM- and placebo-treated patients (P = .21).
Significant increases were noted from baseline to week 24 in patients treated with FCM vs. placebo/saline for haemoglobin (P = .028), ferritin (P ≤ .001), and TSAT levels (P ≤ .001) (Table 2). Serum levels of creatinine, estimated glomerular filtration rate, bilirubin, blood urea nitrogen, aspartate aminotransferase, alanine aminotransferase, gamma-glutamyl transferase, or C-reactive protein were not different between treatment groups at week 24 (all P > .05).
Safety
Survival status was available for all patients through week 52 with no patient having died during the conduct of the trial. A total of 5 of 21 patients receiving placebo/saline and 1 of 18 patients receiving FCM were hospitalized for cardiovascular reasons during the conduct of the trial—in total 8 vs. 2 cardiovascular hospitalization events were observed in the two treatment groups, respectively (P = .045). Nine patients in the placebo group and 3 patients in the FCM group reported at least one serious adverse event (P = .085). The total number of adverse events (76 vs. 114) and serious adverse events (5 vs. 19) was lower in patients treated with FCM compared to placebo. To account for the number of events and observation time per patient, we used a negative binomial regression showing a rate ratio of 0.38 (95% CI: 0.17, 0.88; P = .023) for the number of adverse events and of 0.27 (95% CI: 0.07, 0.96; P = .043) for the number of serious adverse events. Results for serious adverse events were primarily driven by adverse events that required hospitalization or prolongation of an existing hospitalization (16 vs. 4 events) and by events that were deemed life-threatening (3 vs. none). Detailed data on serious and non-serious adverse events are shown in Supplementary data online, Table S3. The study treatment was stopped prematurely in 3 of the 18 patients assigned to receive FCM and in 1 of the 21 patients assigned to receive placebo. No severe allergic reactions related to the study treatment were reported.
Discussion
The results of our trial show for the first time that FCM improves exercise capacity as assessed by the 6MWTD in patients with HFpEF and ID as defined using standard criteria for the diagnosis of ID proposed in the Guidelines of the ESC and American College of Cardiology (ACC)/American Heart Association (AHA)/Heart Failure Society of America (HFSA). The change in 6MWTD with FCM vs placebo by week 24 in the FAIR-HFpEF trial—even though being based on a small number of subjects—suggests presence of a meaningful change that is at least as big or may even go beyond what was seen in the FAIR-HF trial, the first large-scale trial in patients with HFrEF and ID published some 15 years ago, in which the increase was 35 ± 8 m by week 24 in the group that received FCM (Structured Graphical Abstract).4
No significant changes, however, were noted for self-reported PGA of well-being, HRQoL assessments, or NYHA functional class, which is somewhat at odds with the large improvement in 6MWTD, but buttresses the view that our findings require validation in a larger cohort of patients. Importantly, no significant differences were noted for changes in laboratory markers including for biomarkers of kidney and liver function and inflammation between patients receiving FCM or placebo/saline, supporting the safety of FCM therapy. FCM slightly increased haemoglobin values and corrected measures of ID as expected (Structured Graphical Abstract).
It is crucial to recognize two key points. Firstly, the standard criteria for diagnosing ID are applicable in patients with HFpEF, and the treatment of patients with ID using these criteria yields clinical improvement. The debate on this point, however, is far from finished, and a different definition of ID such as TSAT < 20% alone may better identify patients to benefit from intravenous iron treatment.15 Secondly, the potential improvement in 6MWTD achieved through FCM treatment appears to surpass the threshold of clinical relevance, which is estimated at ∼25–35 m with HF or other cardiopulmonary diseases such as pulmonary hypertension.16,17 Notably, a study by Täger et al.18, drawing on data from over 970 patients, suggested that the minimal important difference before and after an intervention should surpass this level. This reflects a 10% enhancement in 6MWTD, particularly noteworthy for individuals with severely limited baseline distances below 400 m, just like in our study cohort of comparatively old subjects whose median age was 80 years, highlighting the importance of treatment effects also for older populations.
A recently published systematic review has underscored the challenges in enhancing exercise capacity among patients with HF.19 In those with HFrEF, many guideline-recommended therapies, including angiotensin-converting enzyme (ACE) inhibitors, sacubitril/valsartan, beta-blockers, mineralocorticoid receptor antagonists, and SGLT2 inhibitors such as dapagliflozin and empagliflozin, have shown limited efficacy in increasing 6MWTD or peak oxygen consumption. Only ivabradine has demonstrated notable benefits.19,20 The situation is similar in HFpEF, where trials of ACE inhibitors and other frequently utilized medications have failed to improve exercise capacity. While the perindopril in elderly people with chronic heart failure (PEP-CHF) trial suggested improvements with perindopril in patients with HFpEF, it fell short of its primary endpoint, complicating the analysis of secondary outcomes.21 Dapagliflozin has shown some modest improvement in 6MWTD (mean effect size of 20.1 m), but similar effects have not been consistently observed with empagliflozin.22,23 Similarly, semaglutide has shown modest improvement in 6MWTD of 14.3 and 21.5 m in patients with HFpEF and obesity with and without diabetes, respectively.24,25 Overall, the significance of enhancing exercise capacity by replenishing iron stores in HFpEF appears to be a promising therapeutic approach.
The loss of a significant treatment effect at 52 weeks may, on the one hand, be related to the reduced number of patients investigated at that time (14 on FCM vs. 12 on placebo) compared to week 24 (15 on FCM vs. 16 on placebo)—which in part is due to the hospitalization events observed mostly in the placebo group—but on the other hand, may also underscore the need for sufficient, repeat iron administration to achieve clinical impact.14 In this regard, the time period of 20 weeks from last administration of FCM at 32–52 weeks may be too long and the cumulative dose of at most 500 mg FCM in the last 6 months prior to the assessment at week 52 too little. In contrast, at the 24-week time point (used for the primary endpoint assessment), the last administration of FCM was only 8 weeks prior to the assessment time (i.e. at week 16) and the cumulative dose FCM in the 6 months prior was 3–5 times higher (i.e. 1500–2500 mg). The only other consistently effective strategy for improving exercise capacity in patients with both HFrEF and HFpEF remains exercise training.26–30
Iron is an essential component of mitochondrial enzymes involved in generating cellular energy in the form of adenosine triphosphate and phosphocreatine in skeletal muscle. Charles-Edwards et al.31 have provided mechanistic insights suggesting that iron repletion therapy improves the phosphocreatine recovery half-time in skeletal muscle of iron-deficient patients with HFrEF. This observation is significant not only for iron-deficient patients, but potentially also for those with sarcopenia, a muscle-wasting syndrome frequently encountered in elderly populations and in patients with HF, likewise contributing to reduced exercise capacity.32–34
We detected statistically significant and clinically meaningful improvements in 6MWTD, but not in measures of HRQoL. In the KCCQ overall summary score, we found a numerical change of 6.5 ± 5.1 points at week 24 (P = .21). We consider these results as promising, but requiring validation. In FAIR-HF (in HFrEF), the same endpoint showed an increase of 7 ± 2 points (P < .001). The trial had a more than 10 times greater number of patients included and hence much more power to detect such a change reliably. The KCCQ is a self-administered, disease-specific questionnaire capturing physical function, symptoms, social function, self-efficacy, and QoL. In HFpEF, there is limited experience with therapies that can positively impact KCCQ. In PARAGON-HF, it was demonstrated that sacubitril/valsartan can somewhat reduce the decline in KCCQ scores over time (1.0 points treatment difference at 12 months). For SGLT2 inhibitors in HFpEF, in the DELIVER and EMPEROR-Preserved studies with dapagliflozin and empagliflozin, respectively, KCCQ results improved by 1–2 points over 8–12 months.11,35 Much more meaningful results were achieved in the recent STEP-HFpEF trials in obese patients with HFpEF with and without diabetes, where semaglutide therapy increased the KCCQ overall summary score on average by 7.5 points within 12 months (P < .0001).36 Larger trials are needed to validate our findings on 6MWTD improvement and to establish whether intravenous iron can improve QoL in patients with HFpEF and ID. They also should be longer in duration to test the intriguing, but very preliminary safety results seen here, suggesting that serious adverse events—and namely hospitalisation rates—may be reduced by medically correcting ID in patients with HFpEF.
Limitations
The study was smaller than originally planned, and the large changes in 6MWTD are based on an overall small sample size. Interestingly, similar effect sizes for 6MWTD have been reported with testosterone in men and women with HFrEF.37,38 Furthermore, the study suffered from some premature study discontinuations and not all patients retained in the study performed the 6MWT at all visits. Clearly, these results require confirmation overall and in key subgroups including those with and without anaemia, renal dysfunction, coronary artery disease, and TSAT < 20%.39
Supplementary Material
Contributor Information
Stephan von Haehling, Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg- August University, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), Partner site Lower Saxony, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany.
Wolfram Doehner, Berlin Institute of Health-Center for Regenerative Therapies (BCRT), Charité- Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353, Berlin, Germany; Deutsches Herzzentrum der Charité, Department of Cardiology (Campus Virchow), German Centre for Cardiovascular Research (DZHK) Partner site Berlin, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353, Berlin, Germany; Center for Stroke Research Berlin, Charité—Universitätsmedizin Berlin, Berlin, Germany.
Ruben Evertz, Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg- August University, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), Partner site Lower Saxony, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany.
Tania Garfias-Veitl, Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg- August University, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany; DZHK (German Center for Cardiovascular Research), Partner site Lower Saxony, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany.
Carlotta Derad, DZHK (German Center for Cardiovascular Research), Partner site Lower Saxony, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany; Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany.
Monika Diek, Deutsches Herzzentrum der Charité, Department of Cardiology (Campus Virchow), German Centre for Cardiovascular Research (DZHK) Partner site Berlin, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353, Berlin, Germany.
Mahir Karakas, Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner site HH/Kiel/HL, Hamburg, Germany.
Ralf Birkemeyer, Herzklinik Ulm, Ulm, Germany.
Gerasimos Fillippatos, Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece.
Mitja Lainscak, Division of Cardiology, General Hospital Murska Sobota, 9000 Murska Sobota, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
Javed Butler, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA; Baylor Scott and White Research Institute, Dallas, TX, United States of America.
Piotr Ponikowski, Center for Heart Diseases, University Hospital, Wroclaw Medical University, Wroclaw, Poland.
Michael Böhm, Department of Internal Medicine Clinic III, Saarland University Hospital, Homburg/Saar, Germany.
Tim Friede, DZHK (German Center for Cardiovascular Research), Partner site Lower Saxony, Robert-Koch-Strasse 40, D-37075, Goettingen, Germany; Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany.
Stefan D Anker, Berlin Institute of Health-Center for Regenerative Therapies (BCRT), Charité- Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353, Berlin, Germany; Deutsches Herzzentrum der Charité, Department of Cardiology (Campus Virchow), German Centre for Cardiovascular Research (DZHK) Partner site Berlin, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353, Berlin, Germany.
Supplementary data
Supplementary data are available at European Heart Journal online.
Declarations
Disclosure of Interest
S.v.H. has been a paid consultant for and/or received honoraria payments from Amomed, AstraZeneca, Bayer, Boehringer Ingelheim, BRAHMS, Edwards Lifesciences, MSD, Novo Nordisk, Novartis, Pfizer, Pharmacosmos, Respicardia, Roche, Servier, Sorin, and Vifor. S.v.H. reports research support from Amgen, AstraZeneca, Boehringer Ingelheim, Pharmacosmos, IMI, and the German Center for Cardiovascular Research (DZHK). W.D. reports consulting fees from Aimediq, Bayer, Boehringer Ingelheim, Boston Scientific, Lilly, Medtronic, Pfizer, Sanofi-Aventis, Sphingotec, Vifor Pharma, travel support from Pharmacosmos, and research support to the Institute from EU (Horizon2020), German Ministry of Education and Research, German Center for Cardiovascular Research, Boehringer Ingelheim, from Vifor Pharma related to this trial, and ZS Pharma. M.K. reports consulting fees and honoraria payments within the last 3 years from Adrenomed, 4TEEN4 Pharmaceuticals, Pharmacosmos, Sphingotec, and CSLVifor, all not related to the submitted work, and research support within the last 3 years from the German Research Foundation (DFG), the German Ministry of Education and Research (BMBF), the German Center for Cardiovascular Research (DZHK), the European Union (Horizon 2020), the Else Kroener-Fresenius Foundation (EKFS Clinician Scientist Professorship), Adrenomed AG, and CSL Vifor, all not related to the submitted work. G.F. reports lecture fees and /or advisory and/or trial committee membership by Bayer, Boehringer Ingelheim, Servier, Novartis, Impulse Dynamics, Vifor, Medtronic, Cardior, Novo Nordisc and Research Grants from the European Union. M.L. reports personal fees from AstraZeneca, Boehringer Ingelheim, Pfizer, and Bayer. J.B. serves as a consultant to Abbott, Adrenomed, Amgen, Array, AstraZeneca, Bayer, Berlin Cures, Boehringer Ingelheim, Bristol-Myers Squib, CVRx, G3 Pharmaceutical, Innolife, Janssen, LivaNova, Luitpold, Medtronic, Merck, Novartis, Novo Nordisk, Occlutech, Relypsa, Roche, Sanofi, SC Pharma, V-Wave Limited, and Vifor. P.P. reports consulting fees from Boehringer Ingelheim, AstraZeneca, Vifor Pharma, Amgen, Servier, Novartis, Bayer, MSD, Pfizer, Cibiem, Impulse Dynamics, Renal Guard Solutions, and BMS and has also received honoraria from Boehringer Ingelheim, AstraZeneca, Vifor Pharma, Amgen, Servier, Novartis, Berlin Chemie, Bayer, Pfizer, Impulse Dynamics, Renal Guard Solutions, BMS, and Abbott Vascular for lectures, presentations, speakers' bureaus, manuscript writing, or educational events. M.B. was supported by the Deutsche Forschungsgemeinschaft (German Research Foundation; TTR 219, project no. 322900939) and reports personal fees from Abbott, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Cytokinetics, Medtronic, Novartis, Servier, and Vifor. T.F. reports personal fees for statistical consultancies (including data monitoring committees) from Actimed, Bayer, BiosenseWebster, BMS, Coherex Medical, CSL Behring, Enanta, Fresenius Kabi, Galapagos, IQVIA, Janssen, KyowaKirin, LivaNova, Minoryx, Novartis, r-connect, Recardio, Relaxera, Roche, Servier, Viatris, and Vifor, all outside the submitted work. S.D.A. reports grants and personal fees from Vifor and Abbott Vascular and personal fees for consultancies, trial committee work and/or lectures from Actimed, Amgen, Astra Zeneca, Bayer, Boehringer Ingelheim, Bioventrix, Brahms, Cardiac Dimensions, Cardior, Cordio, CVRx, Edwards, Farraday, Impulse Dynamics, Janssen, Novartis, Occlutech, Pfizer, Respicardia, Servier, Vectorious, and V-Wave and declares that he is named co-inventor of two patent applications regarding MR-proANP (DE 102007010834 and DE 102007022367), but he does not benefit personally from the related issued patents. All other authors report no conflict of interest.
Data Availability
Requests for data analyses or sharing can be made towards S.v.H. and S.D.A. Such requests should be accompanied by an outline of the proposed study and a detailed statistical analysis plan. Proposals will be considered on a case by case basis, based on their scientific merit and availability of data and resources, but they also need to keep in mind German data protection laws and related requirements.
Funding
The study was funded by Vifor Pharma (Glattbrugg, Switzerland) by providing an unrestricted grant to Charité—Universitätsmedizin Berlin, Germany. The latter is the legal sponsor of the trial.
Ethical Approval
An independent ethics committee approved the protocol at all participating centres, and all subjects provided written informed consent.
Pre-registered Clinical Trial Number
The trial was registered at clinicaltrials.gov (NCT03074591).
References
- 1. Rocha BML, Cunha GJL, Menezes Falcão LF. The burden of iron deficiency in heart failure: therapeutic approach. J Am Coll Cardiol 2018;71:782–93. 10.1016/j.jacc.2017.12.027 [DOI] [PubMed] [Google Scholar]
- 2. Savarese G, von Haehling S, Butler J, Cleland JGF, Ponikowski P, Anker SD. Iron deficiency and cardiovascular disease. Eur Heart J 2023;44:14–27. 10.1093/eurheartj/ehac569 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. von Haehling S, Ebner N, Evertz R, Ponikowski P, Anker SD. Iron deficiency in heart failure: an overview. JACC Heart Fail 2019;7:36–46. 10.1016/j.jchf.2018.07.015 [DOI] [PubMed] [Google Scholar]
- 4. Anker SD, Comin Colet J, Filippatos G, Willenheimer R, Dickstein K, Drexler H, et al. Ferric carboxymaltose in patients with heart failure and iron deficiency. N Engl J Med 2009;361:2436–48. 10.1056/NEJMoa0908355 [DOI] [PubMed] [Google Scholar]
- 5. Ponikowski P, Kirwan B-A, Anker SD, McDonagh T, Dorobantu M, Drozdz J, et al. Ferric carboxymaltose for iron deficiency at discharge after acute heart failure: a multicentre, double-blind, randomised, controlled trial. Lancet 2020;396:1895–904. 10.1016/S0140-6736(20)32339-4 [DOI] [PubMed] [Google Scholar]
- 6. Kalra PR, Cleland JGF, Petrie MC, Thomson EA, Kalra PA, Squire IB, et al. Intravenous ferric derisomaltose in patients with heart failure and iron deficiency in the UK (IRONMAN): an investigator-initiated, prospective, randomised, open-label, blinded-endpoint trial. Lancet 2022;400:2199–209. 10.1016/S0140-6736(22)02083-9 [DOI] [PubMed] [Google Scholar]
- 7. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: developed by the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). With the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail 2022;2:4–131. 10.1002/ejhf.2333 [DOI] [PubMed] [Google Scholar]
- 8. Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, et al. 2022 AHA/ACC/HFSA Guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. J Am Coll Cardiol 2022;79:e263–421. 10.1016/j.jacc.2021.12.012 [DOI] [PubMed] [Google Scholar]
- 9. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, et al. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med 2021;385:1451–61. 10.1056/NEJMoa2107038 [DOI] [PubMed] [Google Scholar]
- 10. von Haehling S, Assmus B, Bekfani T, Dworatzek E, Edelmann F, Hashemi D, et al. Heart failure with preserved ejection fraction: diagnosis, risk assessment, and treatment. Clin Res Cardiol 2024. 10.1007/s00392-024-02396-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Anker SD, Usman MS, Anker MS, Butler J, Böhm M, Abraham WT, et al. Patient phenotype profiling in heart failure with preserved ejection fraction to guide therapeutic decision making. A scientific statement of the Heart Failure Association, the European Heart Rhythm Association of the European Society of Cardiology, and the European Society of Hypertension. Eur J Heart Fail 2023;25:936–55. 10.1002/ejhf.2894 [DOI] [PubMed] [Google Scholar]
- 12. Beale AL, Warren JL, Roberts N, Meyer P, Townsend NP, Kaye D. Iron deficiency in heart failure with preserved ejection fraction: a systematic review and meta-analysis. Open Heart 2019;6:e001012. 10.1136/openhrt-2019-001012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. von Haehling S, Doehner W, Evertz R, Garfias-Veitl T, Diek M, Karakas M, et al. Iron deficiency in heart failure with preserved ejecton fraction: rationale and design of the FAIR-HFpEF trial. Glob Cardiol 2023;1:39–46. 10.4081/cardio.2023.15 [DOI] [Google Scholar]
- 14. Talha KM, Butler J, von Haehling S, Lainscak M, Ponikowski P, Anker SD. Defining iron replete status in patients with heart failure treated with intravenous iron. Glob Cardiol 2023;1:7–11. 10.4081/cardio.2023.17 10.4081/cardio.2023.17 [DOI] [Google Scholar]
- 15. Packer M, Anker SD, Butler J, Cleland JGF, Kalra PR, Mentz RJ, et al. Identification of three mechanistic pathways for iron-deficient heart failure. Eur Heart J 2024;45:2281–93. 10.1093/eurheartj/ehae284 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Rosano GMC, Farkas J. Evolving targets for heart failure: the journey so far. Glob Cardiol 2023;1:3–6. 10.4081/cardio.2023.6 [DOI] [Google Scholar]
- 17. Khan MS, Anker SD, Friede T, Jankowska EA, Metra M, Piña IL, et al. Minimal clinically important differences in 6-minute walk test in patients with HFrEF and iron deficiency. Card Fail 2023;29:760–70. 10.1016/j.cardfail.2022.10.423 [DOI] [PubMed] [Google Scholar]
- 18. Täger T, Hanholz W, Cebola R, Fröhlich H, Franke J, Doesch A, et al. Minimal important difference for 6-minute walk test distances among patients with chronic heart failure. Int J Cardiol 2014;176:94–8. 10.1016/j.ijcard.2014.06.035 [DOI] [PubMed] [Google Scholar]
- 19. von Haehling S, Arzt M, Doehner W, Edelmann F, Evertz R, Ebner N, et al. Improving exercise capacity and quality of life using non-invasive heart failure treatments: evidence from clinical trials. Eur J Heart Fail 2021;23:92–113. 10.1002/ejhf.1838 [DOI] [PubMed] [Google Scholar]
- 20. Volterrani M, Cice G, Caminiti G, Vitale C, D’Isa S, Perrone Filardi P, et al. Effect of carvedilol, ivabradine or their combination on exercise capacity in patients with heart failure (the CARVIVA HF trial). Int J Cardiol 2011;151:218–24. 10.1016/j.ijcard.2011.06.098 [DOI] [PubMed] [Google Scholar]
- 21. Cleland JG, Tendera M, Adamus J, Freemantle N, Polonski L, Taylor J, et al. The perindopril in elderly people with chronic heart failure (PEP-CHF) study. Eur Heart J 2006;27:2338–45. 10.1093/eurheartj/ehl250 [DOI] [PubMed] [Google Scholar]
- 22. Nassif ME, Windsor SL, Borlaug BA, Kitzman DW, Shah SJ, Tang F, et al. The SGLT2 inhibitor dapagliflozin in heart failure with preserved ejection fraction: a multicenter randomized trial. Nat Med 2021;27:1954–60. 10.1038/s41591-021-01536-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Abraham WT, Lindenfeld J, Ponikowski P, Agostoni P, Butler J, Desai AS, et al. Effect of empagliflozin on exercise ability and symptoms in heart failure patients with reduced and preserved ejection fraction, with and without type 2 diabetes. Eur Heart J 2021;42:700–10. 10.1093/eurheartj/ehaa943 [DOI] [PubMed] [Google Scholar]
- 24. Kosiborod MN, Abildstrøm SZ, Borlaug BA, Butler J, Rasmussen S, Davies M, et al. Semaglutide in patients with heart failure with preserved ejection fraction and obesity. N Engl J Med 2023;389:1069–84. 10.1056/NEJMoa2306963 [DOI] [PubMed] [Google Scholar]
- 25. Kosiborod MN, Petrie MC, Borlaug BA, Butler J, Davies M, Hovingh GK, et al. Semaglutide in patients with obesity-related heart failure and type 2 diabetes. N Engl J Med 2023;389:1069–84. 10.1056/NEJMoa2306963 [DOI] [PubMed] [Google Scholar]
- 26. Kitzman DW, Brubaker PH, Morgan TM, Stewart KP, Little WC. Exercise training in older patients with heart failure and preserved ejection fraction: a randomized, controlled, single-blind trial. Circ Heart Fail 2010;3:659–67. 10.1161/CIRCHEARTFAILURE.110.958785 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Edelmann F, Gelbrich G, Düngen H-D, Fröhling S, Wachter R, Stahrenberg R, et al. Exercise training improves exercise capacity and diastolic function in patients with heart failure with preserved ejection fraction: results of the Ex-DHF (Exercise training in Diastolic Heart Failure) pilot study. J Am Coll Cardiol 2011;58:1780–91. 10.1016/j.jacc.2011.06.054 [DOI] [PubMed] [Google Scholar]
- 28. Smart NA, Haluska B, Jeffriess L, Leung D. Exercise training in heart failure with preserved systolic function: a randomized controlled trial of the effects on cardiac function and functional capacity. Congest Heart Fail 2012;18:295–301. 10.1111/j.1751-7133.2012.00295.x [DOI] [PubMed] [Google Scholar]
- 29. Kitzman DW, Brubaker PH, Herrington DM, Morgan TM, Stewart KP, Hundley WG, et al. Effect of endurance exercise training on endothelial function and arterial stiffness in older patients with heart failure and preserved ejection fraction: a randomized, controlled, single-blind trial. J Am Coll Cardiol 2013;62:584–92. 10.1016/j.jacc.2013.04.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Kitzman DW, Brubaker P, Morgan T, Haykowsky M, Hundley G, Kraus WE, et al. Effect of caloric restriction or aerobic exercise training on peak oxygen consumption and quality of life in obese older patients with heart failure with preserved ejection fraction: a randomized clinical trial. JAMA 2016;315:36–46. 10.1001/jama.2015.17346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Charles-Edwards G, Amaral N, Sleigh A, Ayis S, Catibog N, McDonagh T, et al. Effect of iron isomaltoside on skeletal muscle energetics in patients with chronic heart failure and iron deficiency. Circulation 2019;139:2386–98. 10.1161/CIRCULATIONAHA.118.038516 [DOI] [PubMed] [Google Scholar]
- 32. Fülster S, Tacke M, Sandek A, Ebner N, Tschöpe C, Doehner W, et al. Muscle wasting in patients with chronic heart failure: results from the studies investigating co-morbidities aggravating heart failure (SICA-HF). Eur Heart J 2013;34:512–9. 10.1093/eurheartj/ehs381 [DOI] [PubMed] [Google Scholar]
- 33. von Haehling S, Garfias Macedo T, Valentova M, Anker MS, Ebner N, Bekfani T, et al. Muscle wasting as an independent predictor of survival in patients with chronic heart failure. J Cachexia Sarcopenia Muscle 2020;11:1242–9. 10.1002/jcsm.12603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Fonseca GWPD, Garfias Macedo T, Ebner N, Dos Santos MR, de Souza FR, Mady C, et al. Muscle mass, muscle strength, and functional capacity in patients with heart failure of chagas disease and other aetiologies. ESC Heart Fail 2020;7:3086–94. 10.1002/ehf2.12936 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Solomon SD, McMurray JJV, Anand IS, Ge J, Lam CSP, Maggioni AP, et al. Angiotensin-neprilysin inhibition in heart failure with preserved ejection fraction. N Engl J Med 2019;381:1609–20. 10.1056/NEJMoa1908655 [DOI] [PubMed] [Google Scholar]
- 36. Butler J, Shah SJ, Petrie MC, Borlaug BA, Abildstrøm SZ, Davies MJ, et al. Semaglutide versus placebo in people with obesity-related heart failure with preserved ejection fraction: a pooled analysis of the STEP-HFpEF and STEP-HFpEF DM randomised trials. Lancet 2024;403:1635–48. 10.1016/S0140-6736(24)00469-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Caminiti G, Volterrani M, Iellamo F, Marazzi G, Massaro R, Miceli M, et al. Effect of long-acting testosterone treatment on functional exercise capacity, skeletal muscle performance, insulin resistance, and baroreflex sensitivity in elderly patients with chronic heart failure a double-blind, placebo-controlled, randomized study. J Am Coll Cardiol 2009;54:919–27. 10.1016/j.jacc.2009.04.078 [DOI] [PubMed] [Google Scholar]
- 38. Iellamo F, Volterrani M, Caminiti G, Karam R, Massaro R, Fini M, et al. Testosterone therapy in women with chronic heart failure: a pilot double-blind, randomized, placebo-controlled study. J Am Coll Cardiol 2010;56:1310–6. 10.1016/j.jacc.2010.03.090 [DOI] [PubMed] [Google Scholar]
- 39. Ponikowski P, Mentz RJ, Hernandez AF, Butler J, Khan MS, van Veldhuisen DJ, et al. Efficacy of ferric carboxymaltose in heart failure with iron deficiency: an individual patient data meta-analysis. Eur Heart J 2023;44:5077–91. 10.1093/eurheartj/ehad586 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Requests for data analyses or sharing can be made towards S.v.H. and S.D.A. Such requests should be accompanied by an outline of the proposed study and a detailed statistical analysis plan. Proposals will be considered on a case by case basis, based on their scientific merit and availability of data and resources, but they also need to keep in mind German data protection laws and related requirements.