Skip to main content

Effects of ferric citrate hydrate in patients with chronic kidney disease and heart failure: subgroup analysis of a long-term, real-world, post-marketing surveillance study

Abstract

Background

Iron deficiency is widely present in patients with heart failure (HF) and is associated with an increased risk of mortality and poor clinical outcomes regardless of anemia. HF is highly prevalent in patients with chronic kidney disease (CKD). However, existing oral iron preparations have failed to improve iron-related parameters in patients with HF, and intravenous iron preparations are recommended. Ferric citrate hydrate (FC) is an oral iron-based phosphate binder for CKD that is also approved for the treatment of patients with iron-deficiency anemia in Japan. In this subgroup analysis, we evaluated the effect of oral FC on iron-related parameters in CKD patients with and without HF.

Methods

We examined iron- and phosphate-related parameters and adverse drug reactions in subpopulations of CKD patients with and without HF enrolled in a previously reported 104-week, real-world, post-marketing surveillance study of FC in Japan.

Results

Among 2811 enrolled CKD patients, 348 patients had HF and 2352 did not have HF, including 166 and 1401 undergoing hemodialysis (HD), 36 and 173 undergoing peritoneal dialysis (PD), and 146 and 778 non-dialysis-dependent (ND) patients, respectively. The mean changes (95% confidence interval (CI)) in serum ferritin from baseline to week 36 were 90.98 (62.99–118.97) and 81.86 (72.68–91.03) ng/mL in HD, 158.64 (108.91–208.36) and 132.91 (98.59–167.23) ng/mL in PD, and 68.06 (40.40–95.73) and 99.75 (81.10–118.40) ng/mL in ND group, respectively. The mean changes (95% CI) in transferrin saturation (TSAT) (%) from baseline to week 12 in patients with and without HF were 12.79 (9.15–16.44) % and 9.57 (8.46–10.68) % in HD, 9.55 (1.31–17.78) % and 4.96 (1.44–8.48) % in PD, and 5.85 (2.02–9.69) % and 5.21 (3.34–7.09) in ND patients, respectively. Levels of these parameters were well maintained thereafter. Mean serum phosphate levels decreased after FC treatment initiation and were well maintained in all groups.

Conclusions

This study demonstrated that oral FC had a tendency to increase serum ferritin and TSAT, and controlled serum phosphate in CKD patients regardless of the presence of HF.

Trial registration This surveillance was conducted in accordance with the Good Post-marketing Study Practice of Ministry of Health, Labour, and Welfare in Japan.

Background

Patients with heart failure (HF) often have iron deficiency with or without anemia [1, 2]. Previous studies showed a high prevalence of these conditions in patients with acute and chronic HF. Iron deficiency, defined as ferritin < 100 ng/mL or 100–299 ng/mL with transferrin saturation (TSAT) < 20%, was found in 50% of patients with chronic HF [3] and 69–75% of patients with acute HF [4], whereas anemia (defined by the World Health Organization as hemoglobin < 13 g/dL in men and < 12 g/dL in women) was found in 30% of patients with advanced HF [5] and 57–58% of patients with chronic or acute HF [6, 7]. Anemia and iron deficiency are associated with mortality and poor outcomes in HF patients with reduced or preserved left ventricular ejection fraction (HFrEF or HFpEF) [3, 5,6,7,8]. The 2021 European Society of Cardiology (ESC) guidelines recommend periodical screening for anemia and iron deficiency, and the use of intravenous iron preparations with ferric carboxymaltose to treat this condition [1]. Intravenous iron was shown to increase iron-related parameters and improve the exercise capacity and quality of life of patients with chronic HF [9, 10] and to reduce the risk of re-hospitalization in patients who have recovered from acute HF [11]. However, there is currently no evidence to indicate that oral iron preparations can improve outcomes in patients with HF. In the IRONOUT HF randomized trial, oral iron (iron polysaccharide, 150 mg, twice daily for 16 weeks) failed to increase iron absorption (median changes (95% confidence interval (CI)) of serum ferritin and TSAT after 16 weeks of treatment were 18 (− 8 to 38) ng/mL and 2 (− 3 to 7) %, respectively) or improve the exercise capacity or quality of life of patients with HFrEF and iron deficiency [12]. In another randomized study, the erythropoiesis-stimulating agent (ESA) darbepoetin alfa failed to improve mortality and increased the risks of embolic and thrombotic events in patients with systolic HF and mild-to-moderate anemia [13]. ESC guidelines accordingly do not recommend the use of ESAs to treat anemia in patients with HF, and the Japanese Circulation Society guidelines for the treatment of HF state that there is currently no established evidence on how to treat anemia in patients with acute and chronic HF [14, 15].

Ferric citrate hydrate (FC; Riona®; Torii Pharmaceutical Co., Ltd., Tokyo, Japan) has been approved in Japan as an oral iron-based phosphate binder that effectively controls serum phosphate concentrations in patients with chronic kidney disease (CKD), including dialysis- and non-dialysis-dependent patients [16,17,18]. Furthermore, FC increased hemoglobin levels in patients with iron-deficiency anemia with or without CKD [19, 20] and has been approved to treat iron-deficiency anemia in Japan. Ferric citrate (Auryxia®; Akebia Therapeutics, Inc., Cambridge, MA, USA) has the same active ingredient as that of Riona® although the formulation provides different amounts of elemental iron (Riona® has approximately 60 mg elemental iron per 250 mg tablet, whereas Auryxia® has 210 mg elemental iron per 1 g tablet), and it has been approved to treat hyperphosphatemia in patients with dialysis-dependent CKD and iron-deficiency anemia in patients with non-dialysis-dependent CKD in the USA [21, 22]. A previous post hoc analysis compared the iron-related parameters of ferric citrate (Auryxia®) in non-dialysis-dependent patients with CKD and iron-deficiency anemia with or without HF from phase 2 and 3 trials in the USA. They found mean (standard deviation (SD)) increases from baseline to week 12 in serum ferritin of 201.6 (172.3) pmol/L [89.7 (76.7) ng/mL] and TSAT of 10.9 (13.7) % in patients with HF, which were comparable with changes in patients without HF. However, the previous analysis was conducted only in non-dialysis-dependent patients with CKD and iron-deficiency anemia, and the mean ferric citrate (Auryxia®) dose was 5000 mg/day (1050 mg iron/day) [23]. The dose of ferric citrate (Auryxia®) provided a higher dose of elemental iron than the dose used in the IRONOUT HF randomized trial, oral iron polysaccharide (150 mg, twice daily, 300 mg iron/day) [12].

We previously conducted a 104-week, real-world, post-marketing surveillance study of FC in dialysis- and non-dialysis-dependent patients with CKD [24] and showed that FC improved iron-related parameters, with no new safety concerns. In the current study, we analyzed the association of FC on iron- and erythrocyte-related parameters, and CKD-mineral and bone disorder (MBD)-related parameters in CKD patients with and without HF using data from this previous post-marketing surveillance study [24]. The study population was expected to include a large subpopulation of patients with HF, given that CKD and HF frequently coexist [1] and the prevalence of CKD in patients with HF in Japan was reported to be 23% [25].

Methods

Study design

This post-marketing surveillance study was conducted in Japan as part of a risk management plan to ensure the long-term safety of a newly approved drug in a real-world setting. Patients were registered centrally from January 30, 2015. Case report forms were collected from participating institutions for up to a maximum of 2 years. Data collection was terminated on April 30, 2020 [24].

Patients

Patients included in the surveillance study have been described in detail in a previous report [24]. Briefly, CKD patients, including patients undergoing hemodialysis (HD group), peritoneal dialysis (PD group), or no dialysis (ND group), were registered within 14 days from the initiation of FC treatment and followed up prospectively. Patients undergoing combined dialysis (HD plus PD) were excluded from the analyses. Among these registered patients, patients diagnosed with comorbid HF by a physician at baseline were defined as patients with HF, and the other patients were defined as patients without HF in the current analyses. All patients whose case report forms were returned at least once after their first visit were analyzed for safety (safety-analysis set) and patients within this set with available effectiveness data were analyzed for effectiveness (effectiveness-analysis set).

FC treatment

FC (250 mg tablet containing approximately 60 mg of elemental ferric iron) was administered orally three times per day immediately after a meal. The starting dose was 500 mg (1500 mg/day) as recommended in the package insert. The dose was adjusted by physicians according to serum phosphate concentrations or clinical status, with a maximum allowed dose of 6000 mg/day. Concomitant medications (e.g., phosphate binders and iron preparations) were allowed [24].

Evaluation of iron- and erythrocyte-related parameters

Iron- and erythrocyte-related parameters, including serum ferritin, serum iron, TSAT, and hemoglobin, were measured as parameters of special interest in the safety-analysis set at baseline, 4, 12, 16, 24, 28, 36, 52, 76, and 104 weeks after the initiation of FC treatment, and at discontinuation of the treatment. The absolute value of each parameter and the difference from baseline were summarized in each group using descriptive statistics. Time-course changes in serum ferritin, and TSAT were plotted for each group throughout the observation period.

Evaluation of CKD-MBD-related parameters

CKD-MBD-related parameters, including serum phosphate, serum calcium, and intact parathyroid hormone, were measured in the effectiveness-analysis set at baseline, 4, 12, 16, 24, 28, 36, 52, 76, and 104 weeks after the initiation of FC treatment, and at treatment discontinuation. Serum calcium was corrected when serum albumin was < 4.0 g/dL using the formula: corrected calcium [mg/dL] = (absolute value of serum calcium [mg/dL]) + [4 − (serum albumin [g/dL])]. When serum albumin was ≥ 4.0 g/dL, corrected calcium was equal to the absolute serum calcium value. The absolute value of each parameter and the difference from baseline were summarized in each group using descriptive statistics. Time-course changes in serum phosphate were plotted for each group throughout the observation period.

Adverse drug reactions

Adverse drug reactions were recorded in the safety-analysis set using preferred terms from MedDRA Ver. 23.0 and were summarized using descriptive statistics. If a patient experienced an event multiple times, it was recorded as one event.

Statistical analysis

Patient characteristics, iron- and erythrocyte-related parameters and adverse drug reactions were analyzed in the safety-analysis set, and CKD-MBD-related parameters were analyzed in the effectiveness-analysis set. On the basis of data from all Japanese pre-approval clinical studies, we planned to enroll 1000 patients in the HD group, 100 in the PD group, and 500 in the ND group in the post-marketing surveillance study to evaluate long-term safety in the entire CKD population [24]. The current study used the same data set. Mean changes from baseline to each time point and the 95% CI were analyzed for serum ferritin and TSAT at 12, 36, 52, and 104 weeks. All statistical analyses were performed using SAS Ver. 9.4 (SAS Institute Inc., Cary, NC, USA).

Results

Patients

In this observational post-marketing surveillance study, 2811 patients with CKD were registered from 573 institutions [24]. Patients whose case report forms were not returned (n = 76) or who underwent combined dialysis (HD and PD) or did not return after the first visit or did not meet registration criteria were excluded. Among the remaining patients, patients with and without comorbid HF were analyzed in this study (safety-analysis set, n = 348 and n = 2352), comprising 166 and 1401 patients in the HD group, 36 and 173 patients in the PD group, and 146 and 778 patients in the ND group, respectively. The effectiveness-analysis set (n = 303 and n = 2041) comprised 146 and 1232 patients in the HD group, 33 and 150 patients in the PD group, and 124 and 659 patients in the ND group, after excluding 45 and 309 patients from the safety-analysis set, respectively (Fig. 1).

Fig. 1
figure 1

Patient flow: Hemodialysis plus peritoneal dialysis. There are duplicates

Patients with and without comorbid HF characteristics are summarized in Table 1. The mean (standard deviation; SD) age of patients with and without comorbid HF was 68.3 (12.3) and 65.3 (12.7) years in the HD group, 66.4 (13.0) and 62.7 (12.6) years in the PD group, and 68.4 (14.2) and 65.7 (13.5) years in the ND group, respectively. There were more male than female patients in all groups. At baseline, the use of iron preparations (oral or intravenous) in patients with and without comorbid HF was 48/166 patients (28.9%) and 361/1401 patients (25.8%) in the HD group, 3/36 patients (8.3%) and 15/173 patients (8.7%) in the PD group and 12/146 patients (8.2%) and 68/778 patients (8.7%) in the ND group, respectively.

Table 1 Patient characteristics (safety-analysis set)

The daily mean (SD) dose of FC during the study in patients with and without comorbid HF was 1054.5 (516.3) mg and 1082.6 (510.7) mg in the HD group, 951.0 (456.6) mg and 976.4 (509.0) mg in the PD group, and 896.9 (524.1) mg and 845.5 (417.3) in the ND group, respectively.

Iron- and erythrocyte-related parameters

Median (interquartile range (IQR): first quartile, third quartile) serum ferritin levels in patients with and without comorbid HF at baseline were 44.50 (23.70, 94.60) ng/mL and 44.50 (21.10, 88.10) ng/mL in the HD group, 146.00 (58.40, 223.60) ng/mL and 85.60 (51.00, 158.00) ng/mL in the PD group, and 91.00 (53.50, 122.30) and 80.65 (41.00, 154.40) in the ND group, respectively (Table 2). Median serum ferritin levels showed a gradually increasing trend in all groups until around 36 weeks after treatment initiation: the mean changes (95% CI) in serum ferritin in patients with and without comorbid HF from baseline to week 36 were 90.98 (62.99–118.97) ng/mL and 81.86 (72.68–91.03) ng/mL in the HD group, 158.64 (108.91–208.36) ng/mL and 132.91 (98.59–167.23) ng/mL in the PD group, and 68.06 ( 40.40–95.73) and 99.75 (81.10–118.40) ng/mL in the ND group, respectively. The levels became stable thereafter (Tables 2, 3 and Fig. 2).

Table 2 Changes in iron- and erythrocyte-related parameters (safety-analysis set)
Table 3 Changes in serum ferritin and TSAT (safety-analysis set)
Fig. 2
figure 2

Time-course changes in serum ferritin (safety-analysis set). Time-course changes in median serum ferritin. Error bars indicate the first to third quartiles. HD, hemodialysis group; PD, peritoneal dialysis group; ND, non-dialysis-dependent group

Mean (SD) TSAT levels in patients with and without comorbid HF at baseline were 21.34 (13.30) % and 22.04 (11.95) % in the HD group, 28.47 (11.01) % and 30.76 (14.37) % in the PD group, and 23.26 (13.55) % and 28.55 (12.08) % in the ND group, respectively (Table 2), and these also showed an increasing trend until around 12 weeks after treatment initiation and then remained stable in all groups: mean changes (95% CI) in TSAT in patients with and without comorbid HF from baseline to week 12 were 12.79 (9.15–16.44) % and 9.57 (8.46–10.68) % in the HD group, 9.55 (1.31, 17.78) % and 4.96 (1.44, 8.48) % in the PD group, and 5.85 (2.02, 9.69) % and 5.21 (3.34, 7.09) % in the ND group, respectively (Table 2, Table 3 and Fig. 3). Hemoglobin levels were well controlled in all groups (Table 2).

Fig. 3
figure 3

Time-course changes in transferrin saturation (safety-analysis set). Time-course changes in mean transferrin saturation. Error bars indicate the standard deviation. HD, hemodialysis group; PD, peritoneal dialysis group; ND, non-dialysis-dependent group

CKD-MBD-related parameters

Mean (SD) serum phosphate levels in patients with and without comorbid HF at baseline were 6.73 (1.45) mg/dL and 6.56 (1.38) mg/dL in the HD group, 6.62 (1.36) mg/dL and 6.06 (1.35) mg/dL in the PD group, and 5.33 (1.09) mg/dL and 5.34 (1.03) mg/dL in the ND group (Table 4). Mean serum phosphate levels decreased after FC treatment initiation and were then maintained, and the levels in patients with and without comorbid HF at 104 weeks were 5.09 (1.22) mg/dL and 5.40 (1.31) mg/dL in the HD group, 5.34 (1.95) mg/dL and 5.10 (1.16) mg/dL in the PD group, and 4.70 (1.05) mg/dL and 4.90 (1.19) mg/dL in the ND group, respectively. Mean serum phosphate levels were well controlled throughout the study (Table 4 and Fig. 4).

Table 4 Changes in chronic kidney disease—mineral and bone disorder-related parameters (effectiveness-analysis set)
Fig. 4
figure 4

Time-course changes in mean serum phosphate (effectiveness-analysis set). Time-course changes in mean serum phosphate. Error bars indicate standard deviations. HD, hemodialysis group; PD, peritoneal dialysis group; ND, non-dialysis-dependent group

Adverse drug reactions

Adverse drug reactions in patients with and without comorbid HF occurred in 34/166 patients (20.48%) and 289/1401 patients (20.63%) in the HD group, 16/36 patients (44.44%) and 35/173 patients (20.23%) in the PD group, and 20/146 patients (13.70%) and 131/778 patients (16.84%) in the ND group, respectively. All adverse drug reactions observed in at least two patients in either group are summarized in Table 5. The most frequently observed events in patients with and without comorbid HF were diarrhea in 2/166 patients (1.20%) and 54/1401 patients (3.85%) in the HD group, 3/36 patients (8.33%) and 10/173 patients (5.78%) in the PD group, and 6/146 patients (4.11%) and 36/778 patients (4.63%) in the ND group, followed by serum ferritin increased, which was observed in 6/166 patients (3.61%) and 43/1401 patients (3.07%) in the HD group, 4/36 patients (11.11%) and 10/173 patients (5.78%) in the PD group, and 2/146 patients (1.37%) and 29/778 patients (3.73%) in the ND group.

Table 5 Adverse drug reactions observed in at least two patients in either group (safety-analysis set)

Discussion

Because existing oral iron preparations have failed to improve iron-related parameters in patients with HF [12] and HF is highly prevalent in patients with CKD [1], this study evaluated the effects of FC in subgroups of CKD patients with and without comorbid HF. Regardless of the presence of comorbid HF, levels of iron- and erythrocyte-related parameters, including serum ferritin, TSAT, and hemoglobin, showed an increasing trend after the initiation of FC treatment and were then maintained during the 104-week study period. Levels of CKD-MBD-related parameters, including serum phosphate, were also well controlled. The overall time-course changes in these parameters in patients with and without comorbid HF subpopulations were similar. In particular, median changes in serum ferritin from baseline to week 36 in the PD groups were greater than those in the HD and ND groups. Regular monitoring of serum ferritin would be necessary to prevent iron overload in patients undergoing peritoneal dialysis. There was no obvious difference in the frequency of adverse drug reactions in the HF subpopulation compared with the without HF population. In addition, the mean treatment doses of FC in patients with and without comorbid HF were comparable: 1054.5 mg/day (approximately 253.1 mg iron/day) versus 1082.6 mg/day (approximately 259.8 mg iron/day) in the HD group, 951.0 mg/day (approximately 228.2 mg iron/day) versus 976.4 mg/day (approximately 234.3 mg iron/day) in the PD group, and 896.9 mg/day (approximately 215.3 mg iron/day) versus 845.5 mg/day (approximately 202.9 mg iron)/day in the ND group, respectively.

In the IRONOUT HF study, oral polysaccharide iron was administered to patients with HF with reduced ejection fraction and iron deficiency for 16 weeks, and the median increases (95% CI) from baseline were 18 (− 8, 38) ng/mL for serum ferritin and 2 (− 3, 7) % for TSAT [12]. The polysaccharide iron administered in the IRONOUT HF study (150 mg twice daily; 300 mg iron/day) was comparable to the elemental iron dose administered in our study. However, the increases in the iron-related parameters from baseline to 12 weeks after FC treatment were greater in this study compared with the IRONOUT HF study. These data suggest that oral FC might be absorbed in CKD patients regardless of the presence of comorbid HF.

A previous post hoc analysis compared the iron-related parameters of ferric citrate (Auryxia®) in non-dialysis-dependent patients with CKD and iron-deficiency anemia with or without HF [23]. They found mean (SD) increases from baseline to week 12 in serum ferritin of 201.6 (172.3) pmol/L [89.7 (76.7) ng/mL] and TSAT of 10.9 (13.7) % in patients with HF, which were comparable with changes in patients without HF. Our results in the ND group were in line with this previous report; however, the mean FC (Riona®) dose in the ND group of 896.9 mg/day (approximately 215.3 mg iron/day) was lower compared with the mean ferric citrate (Auryxia®) dose of 5000 mg/day (1050 mg iron/day) in the previous study. The difference between studies might be related to the levels of inflammation between CKD patients in the USA and Japan. Compared with the previous study, we evaluated the iron- and erythroid-related parameters based on real-world data in CKD patients with HF in Japan in the ND group as well as the HD and PD groups and obtained data related to long-term (104 weeks) FC (Riona®) treatment.

Anemia or iron deficiency is associated with an increased risk of mortality in patients with CKD and patients with HF [3, 5,6,7,8, 26]. The CKDopps study [26] and subgroup analyses of the CRIC study [27] reported that a lower TSAT was associated with higher risks of mortality and cardiovascular events in non-dialysis-dependent CKD patients. In the current study, FC had a tendency to increase the level of TSAT in CKD patients with and without HF, which may reduce the risks of mortality and cardiovascular events. In a mouse model of CKD, the administration of ferric citrate (Auryxia®) increased the TSAT and serum ferritin levels and improved cardiac markers, including B-type natriuretic peptide, atrial natriuretic peptide, and b-myosin heavy chain, and prolonged survival [28].

This study had important limitations. The case report forms used in this study lacked several important pieces of information, including detailed information on HF (e.g., HFrEF, HFpEF, and the severity of HF, such as the New York Heart Association functional classification, and were not adjusted by an independent committee), levels of hepcidin and CRP, and use of intravenous iron preparations or ESA dosage. In addition, because of an observational real-world study, missing data were observed and approximately 13% of patients with and without HF (n = 45/348 (12.9%) and n = 309/2352 (13.1%), respectively) were lost to follow-up and therefore we did not determine the reasons for dropping out of the study. Thus, the excluded patients might have been more likely to be resistant to iron replacement by the oral iron therapy. Further prospective, randomized, controlled studies are needed to confirm the iron absorption effect of oral FC in patients with HF.

Conclusions

This long-term, real-world, post-marketing surveillance study demonstrated that oral FC administration had a tendency to increase iron-related parameters in CKD patients regardless of the presence of HF.

Availability of data and materials

The datasets generated and/or analyzed during the study are available from the corresponding author upon reasonable request.

References

  1. 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. Eur Heart J. 2021;42:3599–726.

    Article  CAS  Google Scholar 

  2. 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.

    Article  CAS  Google Scholar 

  3. Klip IT, Comin-Colet J, Voors AA, Ponikowski P, Enjuanes C, Banasiak W, et al. Iron deficiency in chronic heart failure: an international pooled analysis. Am Heart J. 2013;165:575–82.

    Article  CAS  Google Scholar 

  4. Cohen-Solal A, Damy T, Terbah M, Kerebel S, Baguet JP, Hanon O, et al. High prevalence of iron deficiency in patients with acute decompensated heart failure. Eur J Heart Fail. 2014;16:984–91.

    Article  CAS  Google Scholar 

  5. Horwich TB, Fonarow GC, Hamilton MA, MacLellan WR, Borenstein J. Anemia is associated 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.

    Article  Google Scholar 

  6. Kajimoto K, Sato N, Takano T, Investigators of the Acute Decompensated Heart Failure Syndromes (ATTEND) Registry. Association between anemia, clinical features and outcome in patients hospitalized for acute heart failure syndromes. Eur Heart J Acute Cardiovasc Care. 2015;4:568–76.

    Article  Google Scholar 

  7. Hamaguchi S, Tsuchihashi-Makaya M, Kinugawa S, Yokota T, Takeshita A, Yokoshiki H, et al. Anemia is an independent predictor of long-term adverse outcomes in patients hospitalized with heart failure in Japan. A report from the Japanese Cardiac Registry of Heart Failure in Cardiology (JCARE-CARD). Circ J. 2009;73:1901–8.

    Article  Google Scholar 

  8. Iorio A, Senni M, Barbati G, Greene SJ, Poli S, Zambon E, et al. Prevalence and prognostic impact of non-cardiac co-morbidities in heart failure outpatients with preserved and reduced ejection fraction: a community-based study. Eur J Heart Fail. 2018;20:1257–66.

    Article  Google Scholar 

  9. 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.

    Article  CAS  Google Scholar 

  10. Ponikowski P, van Veldhuisen DJ, Comin-Colet J, Ertl G, Komajda M, Mareev V, et al. Beneficial effects of long-term intravenous iron therapy with ferric carboxymaltose in patients with symptomatic heart failure and iron deficiency. Eur Heart J. 2015;36:657–68.

    Article  CAS  Google Scholar 

  11. Ponikowski P, Kirwan BA, Anker SD, McDonagh T, Dorobantu M, Drozdz J, AFFIRM-AHF Investigators, 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.

    Article  CAS  Google Scholar 

  12. Lewis GD, Malhotra R, Hernandez AF, McNulty SE, Smith A, Felker GM, et al. Effect of oral iron repletion on exercise capacity in patients with heart failure with reduced ejection fraction and iron deficiency: The IRONOUT HF randomized clinical trial. JAMA. 2017;317:1958–66.

    Article  CAS  Google Scholar 

  13. Swedberg K, Young JB, Anand IS, Cheng S, Desai AS, Diaz R, et al. Treatment of anemia with darbepoetin alfa in systolic heart failure. N Engl J Med. 2013;368:1210–9.

    Article  CAS  Google Scholar 

  14. Tsutsui H, Isobe M, Ito H, Ito H, Okumura K, Ono M, et al. JCS 2017/JHFS 2017 guideline on diagnosis and treatment of acute and chronic heart failure- digest version. Circ J. 2019;83:2084–184.

    Article  Google Scholar 

  15. Tsutsui H, Ide T, Ito H, Kihara Y, Kinugawa K, Kinugawa S, et al. JCS/JHFS 2021 guideline focused update on diagnosis and treatment of acute and chronic heart failure. J Card Fail. 2021;27:1404–44.

    Article  Google Scholar 

  16. Yokoyama K, Akiba T, Fukagawa M, Nakayama M, Sawada K, Kumagai Y, et al. Long-term safety and efficacy of a novel iron-containing phosphate binder, JTT-751, in patients receiving hemodialysis. J Ren Nutr. 2014;24:261–7.

    Article  CAS  Google Scholar 

  17. Yokoyama K, Akiba T, Fukagawa M, Nakayama M, Hirakata H. JTT-751 for treatment of patients with hyperphosphatemia on peritoneal dialysis. Nephron Clin Pract. 2014;128:135–40.

    Article  CAS  Google Scholar 

  18. Yokoyama K, Hirakata H, Akiba T, Fukagawa M, Nakayama M, Sawada K, et al. Ferric citrate hydrate for the treatment of hyperphosphatemia in nondialysis-dependent CKD. Clin J Am Soc Nephrol. 2014;9:543–52.

    Article  Google Scholar 

  19. Komatsu N, Arita K, Mitsui H, Nemoto T, Hanaki K. Efficacy and safety of ferric citrate hydrate compared with sodium ferrous citrate in Japanese patients with iron deficiency anemia: a randomized, double-blind, phase 3 non-inferiority study. Int J Hematol. 2021;114:8–17.

    Article  CAS  Google Scholar 

  20. Komatsu N, Akizawa T, Arita K, Mitsui H, Nemoto T, Hanaki K. The effect of ferric citrate hydrate as an iron replacement therapy in Japanese patients with iron deficiency anemia. Rinsho Ketueki. 2021;62:1583–92 (in Japanese).

    Google Scholar 

  21. Lewis JB, Sika M, Koury MJ, Chuang P, Schulman G, Smith MT, et al. Ferric citrate controls phosphorus and delivers iron in patients on dialysis. J Am Soc Nephrol. 2015;26:493–503.

    Article  Google Scholar 

  22. Fishbane S, Block GA, Loram L, Neylan J, Pergola PE, Uhlig K, et al. Effects of ferric citrate in patients with nondialysis-dependent CKD and iron deficiency anemia. J Am Soc Nephrol. 2017;28:1851–8.

    Article  CAS  Google Scholar 

  23. McCullough PA, Uhlig K, Neylan JF, Pergola PE, Fishbane S. Usefulness of oral ferric citrate in patients with iron-deficiency anemia and chronic kidney disease with or without heart failure. Am J Cardiol. 2018;122:683–8.

    Article  CAS  Google Scholar 

  24. Yokoyama K, Hashimoto T, Okuda Y, Matsumoto Y, Ito K, Yamada R, et al. Safety and effectiveness of ferric citrate hydrate in serum phosphorus management of patients with chronic kidney disease: a long-term, real-world, observational, post-marketing surveillance study. Clin Exp Nephrol. 2022;26:688–99.

    Article  CAS  Google Scholar 

  25. Kawashiro N, Kasanuki H, Ogawa H, Matsuda N, Hagiwara N, Heart Institute of Japan-Department of Cardiology (HIJC) Investigators. Clinical characteristics and outcome of hospitalized patients with congestive heart failure: results of the HIJC-HF registry. Circ J. 2008;72:2015–20.

    Article  Google Scholar 

  26. Guedes M, Muenz DG, Zee J, Bieber B, Stengel B, Massy ZA, et al. Serum biomarkers of iron stores are associated with increased risk of all-cause mortality and cardiovascular events in nondialysis CKD patients, with or without anemia. J Am Soc Nephrol. 2021;32:2020–30.

    Article  CAS  Google Scholar 

  27. Mehta RC, Cho ME, Cai X, Lee J, Chen J, He J, et al. Iron status, fibroblast growth factor 23 and cardiovascular and kidney outcomes in chronic kidney disease. Kidney Int. 2021;100:1292–302.

    Article  CAS  Google Scholar 

  28. Francis C, Courbon G, Gerber C, Neuburg S, Wang X, Dussold C, et al. Ferric citrate reduces fibroblast growth factor 23 levels and improves renal and cardiac function in a mouse model of chronic kidney disease. Kidney Int. 2019;96:1346–58.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank all physicians and investigators who cooperated in this surveillance study. The medical writing support was provided by ASCA Corporation (Osaka, Japan).

Funding

This surveillance study was funded by Japan Tobacco Inc. and Torii Pharmaceutical Co., Ltd.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Data collection and analysis were performed by Kenjiro Murakami, Ryoichi Yamada, and Hiroyuki Susai. The first draft of the manuscript was prepared by Kyoko Ito, and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Kyoko Ito.

Ethics declarations

Ethics approval and consent to participate

This observational post-marketing surveillance study was conducted in accordance with the Good Post-Marketing Study Practice (GPSP) of the Ministry of Health, Labour, and Welfare in Japan. The data were anonymized and collected within general clinical practice, and the requirement for informed consent was therefore waived.

Consent for publication

Not applicable.

Competing interests

Kyoko Ito and Noriaki Nishino are employees of Torii Pharmaceutical Co., Ltd., and Kenjiro Murakami, Ryoichi Yamada, and Hiroyuki Susai are employees of Japan Tobacco Inc.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ito, K., Murakami, K., Yamada, R. et al. Effects of ferric citrate hydrate in patients with chronic kidney disease and heart failure: subgroup analysis of a long-term, real-world, post-marketing surveillance study. Ren Replace Ther 8, 64 (2022). https://doi.org/10.1186/s41100-022-00454-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41100-022-00454-z

Keywords