You are on page 1of 6

Usefulness of Oral Ferric Citrate in Patients With

Iron-Deficiency Anemia and Chronic Kidney Disease


With or Without Heart Failure
D1X XPeter A. McCullough, D2X XMD, MPHa*, D3X XKatrin Uhlig, D4X XMD, MSb, D5X XJohn F. Neylan, D6X XMDb,
D7X XPablo E. Pergola, D8X XMDc, and D9X XSteven Fishbane, D10X XMDd

Patients with chronic inflammatory conditions including chronic kidney disease (CKD)
and heart failure (HF) are undertreated with iron-deficiency anemia (IDA). Progressive
inflammation and reduced iron transport associated with CKD and HF may reduce the
efficacy of oral iron therapy. Oral ferric citrate improves anemia markers in CKD, but its
effects in patients with CKD and concomitant HF have not been described. Patients with
CKD not on dialysis and IDA from a phase 2 and 3 trial were treated with ferric citrate
(n = 190) or placebo (n = 188); patients with HF were identified from medical histories.
Hemoglobin response was defined as a 10.0-g/L increase in hemoglobin. Changes in
hemoglobin, transferrin saturation, ferritin, and serum phosphate from baseline to week
12 and the incidence of adverse events potentially related to HF were evaluated. HF was
reported in 22% (n = 81) of patients. The proportion of patients with hemoglobin response
to ferric citrate treatment did not significantly differ in patients with and without HF
(43% vs 49%, respectively; p = 0.47); changes from baseline in hemoglobin, iron parame-
ters, and serum phosphate were similar. Adverse events potentially related to HF were
noted more frequently in patients with HF (ferric citrate, 23%; placebo, 17%) versus
those without HF (ferric citrate, 12%; placebo, 11%). In conclusion, these results indicate
a potential role for ferric citrate in the treatment of IDA in patients with CKD not on dial-
ysis and concomitant HF. © 2018 The Author(s). Published by Elsevier Inc. This is an
open access article under the CC BY-NC-ND license. (http://creativecommons.org/
licenses/by-nc-nd/4.0/) (Am J Cardiol 2018;122:683688)

Heart failure (HF) and anemia are common comorbid- recipients.12 These results suggest that over-the-counter
ities associated with a poor prognosis in patients with iron polysaccharide ineffectively treats IDA in patients
chronic kidney disease (CKD).15 Normal iron absorption with HF, possibly because poor tolerability limits elemental
and transport are impaired in CKD and HF due to associ- iron dose delivery. Ferric citrate, an oral phosphate binder
ated inflammation, and in the case of CKD, by way of approved in adult patients with CKD on dialysis, delivers
changes in hepcidin610; therefore, both CKD and HF are 210 mg of elemental ferric iron per gram.13 Ferric citrate
independently associated with iron-deficiency anemia was recently approved as an iron-replacement agent for the
(IDA).5,11 Effective IDA treatment is especially important treatment of IDA in adult patients with CKD not on dialy-
for patients with CKD and concomitant HF because hemo- sis.13 In 2 randomized clinical trials among patients with
globin (Hgb) concentrations may decrease further due to CKD not on dialysis and IDA, ferric citrate increased Hgb
iron restriction and volume overload.5,9,11 In the recent and transferrin saturation (TSAT) and decreased serum
Oral Iron Repletion Effects on Oxygen Uptake in Heart phosphate levels compared with placebo.14,15 The impact
Failure (IRONOUT-HF)study, patients with HF (n = 225) of ferric citrate treatment on patients with CKD, IDA, and
receiving oral iron polysaccharide (100 mg elemental iron/ HF has not been previously reported. Here, we present the
day) experienced minimal increases in iron stores and exer- results of a hypothesis-generating pooled post hoc analysis
cise/functional capacity compared with placebo of these 2 trials stratified by HF status.
a
Baylor University Medical Center, Baylor Heart and Vascular Hospi-
tal, Baylor Heart and Vascular Institute, Dallas, Texas 75246; bKeryx Bio-
Methods
pharmaceuticals, Inc., Boston, Massachusetts; cRenal Associates PA, San The corresponding author had complete access to all the
Antonio, Texas; and dHofstra Northwell School of Medicine, Great Neck, raw data and takes responsibility for data integrity and the
New York. Manuscript received February 21, 2018; revised manuscript data analyses. The data that support the findings of this
received and accepted April 30, 2018.
study are available from the corresponding author upon rea-
This study was presented in part as a poster at the Twenty-First Annual
Scientific Meeting of the Heart Failure Society of America (HFSA) held at sonable request.
Dallas, TX from September 16, 2017 to September 19, 2017. Individual patient data were pooled from the randomized
See page 687 for disclosure information. periods of a phase 2 (ClinicalTrials.gov identifier:
*Corresponding author: Tel.: (214) 820-7997; fax: (214) 820-7553 NCT01736397) and phase 3 clinical trial (NCT02268994),
E-mail address: peteramccullough@gmail.com (P.A. McCullough). both of which evaluated ferric citrate versus placebo in

0002-9149/© 2018 The Author(s). Published by Elsevier Inc. This is an open access article www.ajconline.org
under the CC BY-NC-ND license. (http://creativecommons.org/licenses/by-nc-nd/4.0/)
https://doi.org/10.1016/j.amjcard.2018.04.062
684 The American Journal of Cardiology (www.ajconline.org)

Table 1
Baseline characteristics (safety analysis set)
Parameter Patients with heart failure (n = 81) Patients without heart failure (n = 297)
Ferric citrate (n = 40) Placebo (n = 41) Ferric citrate (n = 150) Placebo (n = 147)
Age (years) 65.0 (3687) 67.5 (3493) 65.6 (2586) 64.0 (2189)
Women 48% 61% 71% 61%
Black/African-American 35% 22% 27% 26%
Chronic kidney disease stage at baseline
3 40% 42% 41% 48%
4 45% 49% 47% 43%
5 15% 10% 12% 10%
eGFR (mL/min/1.73 m2) 23.8 (850) 25.2 (1051) 27.5 (666) 27.3 (863)
Weight (kg) 99.0 (57.5152.8) 95.8 (58.3157.9) 90.8 (46.9154.7) 89.8 (47.9167.8)
Diabetes mellitus 78% 66% 67% 71%
Ischemic heart disease 38% 61% 25% 20%
Hemoglobin (g/L) 106 (91124) 105 (86125) 104 (85123) 105 (84147)
Transferrin saturation (%) 19.8 (1136) 19.5 (443) 21.1 (650) 20.5 (745)
Ferritin (pmol/L) 224.5 (40.4705.6) 241.6 (31.5582.0) 219.1 (6.7853.9) 198.6 (13.5797.7)
Phosphorus (mmol/L) 1.4 (1.02.8) 1.4 (1.02.0) 1.4 (0.92.4) 1.4 (0.92.2)
Median iPTH (pg/mL) (range) 129.5 (30453) 108.0 (21374) 99.5 (6586) 121.0 (16551)
Median iFGF23 (pg/mL) (IQR) 227.6 (235.8) 171.4 (108.0) 135.5 (132.3) 138.0 (155.9)
Median cFGF23 (RU/mL) (IQR) 544.6 (531.6) 425.7 (545.3) 344.2 (353.7) 331.0 (378.6)
Data are mean (range) unless otherwise indicated.
cFGF23 = C-terminal fibroblast growth factor 23; eGFR = estimated glomerular filtration rate; iFGF23 = intact fibroblast growth factor 23; iPTH = intact
parathyroid hormone; IQR = interquartile range.

patients with IDA and stage 3 to 5 nondialysis-depen- the co-primary end points were changes in TSAT and
dent CKD (estimated glomerular filtration rate <60 mL/ serum phosphate from baseline; in the phase 3 trial, the
min/1.73 m2 by the 4-variable Modification of Diet in primary end point was the proportion of patients achieving
Renal Disease Study equation). The design of these trials a 1.0-g/dL Hgb increase at any time during the random-
has previously been reported (Supplementary Table 1).14,15 ized period.
Both trials were randomized, double-blind, multicenter For the purposes of the pooled post hoc analysis, patients
studies with similar patient populations and drug doses; with baseline HF were identified from diagnoses entered for
however, they differed with respect to the randomization their medical history. The safety analysis set comprised all
period (12 and 16 weeks for the phase 2 and 3 trials, randomized patients who received 1 dose of study medi-
respectively) and ferric citrate dose titration (to achieve cation; the intention-to-treat analysis set comprised patients
target serum phosphate levels of 0.971.13 mmol/L in the who received 1 dose of study medication and had 1
phase 2 trial and a target Hgb increase of 10.0 g/L above baseline and 1 postbaseline laboratory assessment (Sup-
baseline in the phase 3 trial). Also, as inclusion criteria for plementary Figure 1).
baseline serum phosphate levels differed in the phase 2 The primary end point of this analysis was the propor-
and 3 trials (1.29 to <1.94 mmol/L and 1.13 mmol/L, tion of Hgb responders, defined as patients who achieved
respectively), patients in the phase 2 study had higher a 10.0-g/L Hgb increase at any time during the random-
serum phosphate levels at baseline. In the phase 2 trial, ized periods. Mean changes in Hgb, TSAT, ferritin, and

Figure 1. Maximum change in hemoglobin by HF status (intention-to-treat analysis set). Cumulative frequency of the maximum change from baseline in
hemoglobin by HF status. HF = heart failure.
Miscellaneous/Ferric Citrate, IDA, and Heart Failure 685

Table 2
Mean change in hemoglobin, iron parameters, and phosphate from baseline to week 12 (intention-to-treat analysis set)
Variable Patients with heart failure Patients without heart failure
Ferric citrate (n = 40) Placebo (n = 41) Ferric citrate (n = 150) Placebo (n = 147)
Hemoglobin (g/L) 4.1 § 7.3 2.1 § 6.4 5.7 § 8.5 1.1 § 8.6
Transferrin saturation (%) 10.9 § 13.7 1.3 § 7.7 12.0 § 15.2 1.4 § 6.4
Ferritin (pmol/L) 201.6 § 172.3 27.4 § 66.5 210.8 § 198.4 7.4 § 86.1
Serum phosphate (mmol/L) 0.17 § 0.26 0.12 § 0.22 0.15 § 0.23 0.05 § 0.21
Data are mean § standard deviation.

phosphate from baseline to week 12 during the randomized reported in their medical history. Baseline characteristics
period were also evaluated. The maximum change in Hgb were relatively similar across studies and treatment groups
(i.e., the difference between baseline and the highest Hgb (Table 1). Patients in the HF subgroups had a slightly lower
value during follow-up) was also assessed. estimated glomerular filtration rate and a higher rate of
Adverse events (AEs) potentially related to HF were ischemic heart disease than those without HF. Mean base-
considered AEs of special interest, which are listed in Sup- line intact and c-terminal fibroblast growth factor 23
plementary Table 2. (FGF23)—risk markers for mortality in CKD—were higher
Descriptive analyses of baseline characteristics and AEs in patients with HF versus those without HF, respectively
were conducted using the safety analysis set. Analyses of (intact FGF23, 381.5 vs 317.9 pg/mL; c-terminal FGF23,
efficacy with changes in laboratory parameters over time 782.4 vs 519.7 RU/mL).
employed the intention-to-treat analysis set. To assess if the The mean ferric citrate dose in the pooled patient popu-
effect of treatment on Hgb response differed for those with lation was 5.0 g/day (delivering 1,050 mg of elemental
and without HF, treatment-by-HF interaction was tested in iron/day). HF did not modify the treatment effect of ferric
a logistic regression model with responder status as the out- citrate compared with placebo. Among ferric citrate-treated
come and study, HF status, treatment, and treatment-by-HF patients, the frequency of Hgb responders was 43% for
status interaction as predictors. The maximum change in those with HF and 49% for those without HF (p = 0.472 for
Hgb was computed for each patient, and the cumulative fre- treatment-by-HF interaction) (Supplementary Table 3).
quency of the maximum changes was plotted according to Although longer treatment duration resulted in a higher
HF status and treatment group. overall response rate in the 16-week phase 3 study com-
Both trials were conducted in accordance with the Interna- pared with the 12-week phase 2 study, the treatment-by-
tional Conference on Harmonization Good Clinical Practice interaction terms within each study were not statistically
guidelines, the Declaration of Helsinki, and applicable local significant (phase 3 study, p = 0.430; phase 2 study,
regulatory requirements and laws. All versions of the study p = 0.943) (Supplementary Table 3). In the pooled analysis,
protocol were reviewed and approved by the Liberty Institu- mean changes from baseline to 12 weeks in Hgb, iron
tional Review Board (DeLand, Florida and Columbia, Mary- parameters, and serum phosphate were similar among all
land) before patient enrollment at each site. Institutional patients who received ferric citrate, regardless of HF status
review board-approved written informed consent was obtained (Table 2).
from each patient before performing any study assessments. The maximum change from baseline in Hgb was greater
in the ferric citrate groups than in the placebo groups but
similar among patients with and without HF (Figure 1).
Results There was no statistically significant difference
Overall, 190 patients were treated with ferric citrate and (p = 0.5842) in the average time to maximum Hgb change
188 with placebo. A total of 81 patients (22%) had HF among ferric citrate-treated patients with HF (60.1 days)

Table 3
Adverse events of special interest in patients in the pooled analysis with and without heart failure
Patients with heart failure Patients without heart failure
Ferric citrate (n = 40) Placebo (n = 41) Ferric citrate (n = 150) Placebo (n = 147)
AE related to a heart failure safety signal* 9 (23%) 7 (17%) 18 (12%) 16 (11%)
AE related to ischemic heart diseasey 1 (3%) 1 (2%) 1 (1%) 3 (2%)
AE related to cerebrovascular disorderz 0 0 0 0
AE related to either a heart failure safety 10 (25%) 7 (17%) 19 (13%) 16 (11%)
signal, ischemic heart disease, or cerebrovascular disorderx
*Includes preferred terms under SMQ codes for cardiac arrhythmias, heart failure, cardiomyopathy, hyponatremia/syndrome of inappropriate antidiuretic
hormone secretion, hypotonichyporesponsive episode, and pulmonary hypertension.
AE = adverse event; SMQ = Standardized Medical Dictionary for Regulatory Activities Queries.
y
Includes preferred terms under SMQ codes for ischemic heart disease.
z
Includes preferred terms under SMQ codes for cerebrovascular disorders.
x
Includes preferred terms under all SMQ codes above.
686 The American Journal of Cardiology (www.ajconline.org)

Ferric citrate-treated patients had higher AE rates than


placebo recipients. Most treatment-emergent AEs were
mild or moderate across all groups. In both the ferric citrate
and placebo groups, the incidence and severity of treat-
ment-emergent AEs were similar among patients with or
without HF. However, ferric citrate-treated patients with
HF showed the highest incidences of drug-related treat-
ment-emergent AEs, serious AEs, and treatment-emergent
AEs resulting in discontinuation (Supplementary Table 4).
Gastrointestinal AEs were the most common treatment-
emergent AEs among ferric citrate-treated patients and
were similar among patients with and without HF (Supple-
mentary Table 5). Hypophosphatemia was reported as an
AE in 1% and 2% of patients who received ferric citrate
and placebo, respectively.
AEs of special interest among patients with HF are
shown in Table 3. The frequency of AEs possibly related to
HF was higher in patients with HF than in those without HF
(Table 3).

Discussion
This pooled post hoc analysis shows that ferric citrate
effectively delivered iron and improved IDA in patients
with CKD both with and without HF. While these results
are consistent with previous studies among patients with
IDA and HF,16,17 they differ from those of the IRONOUT
HF study, in which oral iron polysaccharide showed mini-
mal increases in TSAT and ferritin and no improvement in
exercise capacity versus placebo.12 However, it is difficult
to draw direct parallels between our study and the IRON-
OUT-HF study because of differences in the eligibility cri-
teria used to define IDA among patients with HF alone
(IRONOUT-HF) versus those with HF and CKD (current
pooled analysis).18 In addition, baseline Hgb levels were
higher in IRONOUT-HF (mean 126 g/L) compared with
the current analysis (mean 104 g/L). Yet, increases in iron
parameters with ferric citrate (TSAT, +11% to 12%; ferri-
tin, +200 pmol/L) were notable when compared with iron
polysaccharide (TSAT, +2%; ferritin, +40 pmol/L), which
could be attributable to the greater elemental iron dose with
ferric citrate (1050 mg/day) versus iron polysaccharide
(100 mg/day). The formulation of the ferric citratecoordi-
nation complex given with food allows for a large amount
of ferric iron to be tolerated and effectively delivered
through the gut, despite the additive inhibitions and iron
restrictions that occur in patients with both HF and CKD;
Figure 2. Mean hemoglobin, transferrin saturation, ferritin, and serum
this distinguishes ferric citrate from other oral iron
phosphate levels by week and HF status (intention-to-treat analysis set).* preparations.14,18
Mean (standard error) changes in (A) hemoglobin, (B) transferrin satura- Intravenous (IV) iron (ferric carboxymaltose) in patients
tion, (C) ferritin, and (D) serum phosphate levels by week and HF status. with HF improves exercise tolerance and quality of life
*Data for study weeks 14 and 16 are from patients in the phase 3 study while reducing the risk of hospitalizations for HF and mor-
only. BL = baseline; HF = heart failure. tality.19,20 Based on results from the Ferinject Assessment
in Patients With Iron Deficiency and Chronic Heart Failure
and Ferric Carboxymaltose Evaluation on Performance in
Patients With Iron Deficiency in Combination With
and without HF (63.4 days). Over the entire randomized Chronic Heart Failure trials,21,22 which defined iron defi-
study periods, mean values for Hgb, TSAT, ferritin, and ciency with the same criteria as the IRONOUT-HF study,
serum phosphate were similar in ferric citrate-treated IV iron is now recommended in the American College of
patients with and without HF. They were also similar Cardiology/American Heart Association guidelines for the
among placebo recipients with and without HF (Figure 2). treatment of patients with New York Heart Association
Miscellaneous/Ferric Citrate, IDA, and Heart Failure 687

functional class IIbIII HF with anemia.23,24 Of note, nei- demonstrated absence of iron stores in patients with normal
ther the Ferinject Assessment in Patients With Iron Defi- ferritin concentrations, suggesting that ferritin may be a
ciency and Chronic Heart Failure nor Ferric poor measure of IDA in patients with HF.29 Therefore, eli-
Carboxymaltose Evaluation on Performance in Patients gibility criteria for prospective trials may define IDA in the
With Iron Deficiency in Combination With Chronic Heart context of other parameters (i.e., Hgb and TSAT) that may
Failure trials were powered to evaluate the effect of IV iron be more relevant to patients with HF. Anemia- and HF-
on major cardiovascular outcomes in anemic versus nona- related quality of life, exercise capacity, freedom from
nemic patients.1,24 transfusion, and the need for rescue therapy with IV iron
Despite these recommendations, the percentage of should be considered as secondary points of interest.
patients with HF who are evaluated and treated for iron Our findings suggest a potential role for ferric citrate-
deficiency remains low in clinical practice.25 This may be a based oral iron treatment of IDA in patients with nondial-
result of the lack of long-term safety and efficacy data or ysis-dependent CKD and HF. Administration of oral ferric
the heterogeneity of guidelines for the diagnosis and man- citrate may not only treat IDA and its symptoms but could
agement of iron deficiency in patients with HF.26 Practical potentially influence HF-specific outcomes in patients with
barriers to providing IV iron therapy may be another reason both conditions.
for the low guideline uptake. An oral iron formulation that
is effective in treating IDA in patients with HF would be
Acknowledgment
attractive given the ease of use and avoidance of parenteral
infusions. Statistical support was provided by Prometrika, LLC and
In our analysis, the most common AEs with ferric citrate funded by Keryx Biopharmaceuticals, Inc. Medical writing
were gastrointestinal symptoms; in the phase 3 trial, 26% support was provided by Meghan Sullivan, PhD, and Vasu-
of patients experienced gastrointestinal AEs of any grade. pradha Vethantham, PhD, of inScience Communications,
For reference, in a systematic review of oral iron supple- Springer Healthcare (New York, New York), and was
ments across 111 trials (not including ferric citrate), the funded by Keryx Biopharmaceuticals, Inc.
reported incidence of gastrointestinal AEs ranged from 4%
to 43% and was directly related to the daily elemental iron
dose; however, the maximum elemental iron dose in the tri- Disclosures
als included in this analysis did not exceed 175 mg/day.27 Dr. McCullough has served as a consultant for and has
Patients with HF experienced more AEs of special interest received research support from Keryx Biopharmaceuticals,
compared with patients without HF, confirming the greater Inc. Drs. Uhlig and Neylan are employees of Keryx Bio-
event risk in patients with HF and concomitant kidney dis- pharmaceuticals, Inc. Dr. Fishbane has served as a consul-
ease and its attendant comorbidities. Ferric citrate also tant for AstraZeneca and Keryx, is on the advisory board
resulted in significant decreases in FGF23 in the overall for Keryx, has received research funding from AstraZe-
study population, although it was not analyzed in patients neca, Corvidia, Fibrogen, and Keryx, and has received hon-
with concomitant HF.14,15 FGF23, a phosphate-regulating oraria from AstraZeneca and Keryx. Dr. Pergola has served
hormone, is known to be a risk factor for cardiovascular as a consultant for Akebia, Alnylam, ExThera, Keryx, and
events in patients with CKD.28 Reata.
This analysis has limitations. Although the current anal-
ysis used prospectively collected data, it was retrospective
in nature. Patients with HF were identified post hoc based Funding
on spontaneously reported HF diagnoses from their past Funding was provided by Keryx Biopharmaceuticals,
medical history, rather than a systematic ascertainment of Inc. (Boston, Massachusetts).
HF status; therefore, left ventricular ejection fraction, func-
tional class, and etiology of HF were not available. The 2
studies, while similar in patient populations and treatment Supplementary Data
doses, had different primary end points and dose-titration Supplementary data associated with this article can be
strategies. Subgroup analysis by race and gender was pre- found, in the online version, at https://doi.org/10.1016/j.amj
cluded by small patient numbers and neither study was card.2018.04.062.
powered for subgroup analysis. Measurement of key (and
potentially informative) biomarkers such as hepcidin-25
was lacking, and outcomes of particular relevance to HF, 1. Goodnough TL, Comin-Colet J, Leal-Noval S, Ozawa S, Takere J,
such as quality of life and exercise capacity, were not Henry D, Javidroozi M, Hohmuth B, Bisbe E, Gross I, Shander A.
assessed. Management of anemia in patients with congestive heart failure. Am J
Due to the limitations mentioned previously, this analy- Hematol 2017;92:88–93.
2. Jankowska EA, Kasztura M, Sokolski M, Bronisz M, Nawrocka S,
sis should be considered as hypothesis-generating; an ade- Oleskowska-Florek W, Zymlinski R, Biegus J, Siwolowski P, Bana-
quately designed and sufficiently powered trial is needed to siak W, Anker SD, Filippatos G, Cleland JG, Ponikowski P. Iron defi-
confirm if ferric citrate treatment is safe and beneficial in ciency defined as depleted iron stores accompanied by unmet cellular
patients with IDA and HF regarding the outcomes of hospi- iron requirements identifies patients at the highest risk of death after
an episode of acute heart failure. Eur Heart J 2014;35:2468–2476.
talization for HF and cardiovascular death. The diagnosis 3. Yilmaz MI, Stenvinkel P, Sonmez A, Saglam M, Yaman H, Kilic S,
of IDA in patients with HF can be challenging. In a study Eyileten T, Caglar K, Oguz Y, Vural A, Cakar M, Altun B, Yenicesu
of patients hospitalized for HF, bone marrow examinations M, Carrero JJ. Vascular health, systemic inflammation and progressive
688 The American Journal of Cardiology (www.ajconline.org)

reduction in kidney function; clinical determinants and impact on car- systolic heart failure: a meta-analysis of randomized controlled trials.
diovascular outcomes. Nephrol Dial Transpl 2011;26:3537–3543. Eur J Heart Fail 2016;18:786–795.
4. Babitt JL, Lin HY. Mechanisms of anemia in CKD. J Am Soc Nephrol 20. Qian C, Wei B, Ding J, Wu H, Wang Y. The efficacy and safety of iron
2012;23:1631–1634. supplementation in patients with heart failure and iron deficiency: a
5. Jankowska EA, Malyszko J, Ardehali H, Koc-Zorawska E, Banasiak systematic review and meta-analysis. Can J Cardiol 2016;32:151–
W, von Haehling S, Macdougall IC, Weiss G, McMurray JJ, Anker 159.
SD, Gheorghiade M, Ponikowski P. Iron status in patients with chronic 21. Anker SD, Comin Colet J, Filippatos G, Willenheimer R, Dickstein K,
heart failure. Eur Heart J 2013;34:827–834. Drexler H, Luscher TF, Bart B, Banasiak W, Niegowska J, Kirwan
6. Dick SA, Epelman S. Chronic heart failure and inflammation: what do BA, Mori C, von Eisenhart Rothe B, Pocock SJ, Poole-Wilson PA,
we really know. Circ Res 2016;119:159–176. Ponikowski P. Ferric carboxymaltose in patients with heart failure and
7. Panwar B, Gutierrez OM. Disorders of iron metabolism and anemia in iron deficiency. N Engl J Med 2009;361:2436–2448.
chronic kidney disease. Semin Nephrol 2016;36:252–261. 22. Ponikowski P, van Veldhuisen DJ, Comin-Colet J, Ertl G, Komajda M,
8. Ganz T. Molecular control of iron transport. J Am Soc Nephrol Mareev V, McDonagh T, Parkhomenko A, Tavazzi L, Levesque V,
2007;18:394–400. Mori C, Roubert B, Filippatos G, Ruschitzka F, Anker SD. Beneficial
9. Ganz T, Nemeth E. Hepcidin and iron homeostasis. Biochim Biophys effects of long-term intravenous iron therapy with ferric carboxymal-
Acta 2012;1823:1434–1443. tose in patients with symptomatic heart failure and iron deficiency.
10. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK, Eur Heart J 2015;36:657–668.
Ganz T. IL-6 mediates hypoferremia of inflammation by inducing the 23. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr., Colvin MM,
synthesis of the iron regulatory hormone hepcidin. J Clin Invest Drazner MH, Filippatos GS, Fonarow GC, Givertz MM, Hollenberg
2004;113:1271–1276. SM, Lindenfeld J, Masoudi FA, McBride PE, Peterson PN, Stevenson
11. Zumbrennen-Bullough K, Babitt JL. The iron cycle in chronic kidney LW, Westlake C. ACC/AHA/HFSA focused update of the
disease (CKD): from genetics and experimental models to CKD 2013 ACCF/AHA Guideline for the Management of Heart Failure: a
patients. Nephrol Dial Transpl 2014;29:263–273. report of the American College of Cardiology/American Heart Associ-
12. Lewis GD, Malhotra R, Hernandez AF, McNulty SE, Smith A, Felker ation Task Force on Clinical Practice Guidelines and the Heart Failure
GM, Tang WHW, LaRue SJ, Redfield MM, Semigran MJ, Givertz Society of America. J Am Coll Cardiol 2017;70:776–803.
MM, Van Buren P, Whellan D, Anstrom KJ, Shah MR, Desvigne- 24. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JG, Coats
Nickens P, Butler J, Braunwald E. Effect of oral iron repletion on exer- AJ, Falk V, Gonzalez-Juanatey JR, Harjola VP, Jankowska EA,
cise capacity in patients with heart failure with reduced ejection frac- Jessup M, Linde C, Nihoyannopoulos P, Parissis JT, Pieske B,
tion and iron deficiency: the IRONOUT HF randomized clinical trial. Riley JP, Rosano GM, Ruilope LM, Ruschitzka F, Rutten FH, van
JAMA 2017;317:1958–1966. der Meer P. ESC Guidelines for the diagnosis and treatment of
13. AURYXIA (ferric citrate) tablets: US prescribing information. Boston, acute and chronic heart failure: the Task Force for the diagnosis
MA: Keryx Biopharmaceuticals, Inc.; 2016. and treatment of acute and chronic heart failure of the European
14. Block GA, Fishbane S, Rodriguez M, Smits G, Shemesh S, Pergola Society of Cardiology (ESC). Developed with the special contribu-
PE, Wolf M, Chertow GM. A 12-week, double-blind, placebo-con- tion of the Heart Failure Association (HFA) of the ESC. Eur J
trolled trial of ferric citrate for the treatment of iron deficiency anemia Heart Fail 2016;18:891–975.
and reduction of serum phosphate in patients with CKD stages 3-5. Am 25. Wienbergen H, Pfister O, Hochadel M, Michel S, Bruder O, Remppis
J Kidney Dis 2015;65:728–736. BA, Maeder MT, Strasser R, von Scheidt W, Pauschinger M, Senges J,
15. Fishbane S, Block GA, Loram L, Neylan J, Pergola PE, Uhlig K, Chertow Hambrecht R. RAID-HF (Registry Analysis of Iron DeficiencyHeart
GM. Effects of ferric citrate in patients with nondialysis-dependent CKD Failure) REGISTRY Study Group. Usefulness of iron deficiency cor-
and iron deficiency anemia. J Am Soc Nephrol 2017;28:1851–1858. rection in management of patients with heart failure [from the Registry
16. Beck-da-Silva L, Piardi D, Soder S, Rohde LE, Pereira-Barretto AC, Analysis of Iron Deficiency-Heart Failure (RAID-HF) Registry]. Am J
de Albuquerque D, Bocchi E, Vilas-Boas F, Moura LZ, Montera MW, Cardiol 2016;118:1875–1880.
Rassi S, Clausell N. IRON-HF study: a randomized trial to assess the 26. Peyrin-Biroulet L, Williet N, Cacoub P. Guidelines on the diagnosis
effects of iron in heart failure patients with anemia. Int J Cardiol and treatment of iron deficiency across indications: a systematic
2013;168:3439–3442. review. Am J Clin Nutr 2015;102:1585–1594.
17. Niehaus ED, Malhotra R, Cocca-Spofford D, Semigran M, Lewis GD. 27. Wahl P, Wolf M. FGF23 in chronic kidney disease. Adv Exp Med Biol
Repletion of iron stores with the use of oral iron supplementation in 2012;728:107–125.
patients with systolic heart failure. J Card Fail 2015;21:694–697. 28. Wolf M. Update on fibroblast growth factor 23 in chronic kidney dis-
18. Fishbane S, Spinowitz B. Update on anemia in ESRD and earlier stages ease. Kidney Int 2012;82:737–747.
of CKD: core curriculum 2018. Am J Kidney Dis 2018;71:423–435. 29. Nanas JN, Matsouka C, Karageorgopoulos D, Leonti A, Tsolakis E,
19. Jankowska EA, Tkaczyszyn M, Suchocki T, Drozd M, von Haehling S, Drakos SG, Tsagalou EP, Maroulidis GD, Alexopoulos GP, Kanakakis
Doehner W, Banasiak W, Filippatos G, Anker SD, Ponikowski P. JE, Anastasiou-Nana MI. Etiology of anemia in patients with advanced
Effects of intravenous iron therapy in iron-deficient patients with heart failure. J Am Coll Cardiol 2006;48:2485–2489.

You might also like