Background: Although renal anemia has attracted widespread attention, a large proportion of chronic kidney disease (CKD) patients with anemia still do not meet the hemoglobin (Hb) targets. The discovery of prolyl hydroxylase domain (PHD) enzymes as regulators of hypoxia-inducible factor (HIF)-dependent erythropoiesis has led to the development of novel therapeutic agents for renal anemia. Roxadustat, the first small-molecule HIF-PHD inhibitor, has completed the phase 3 trials. There are currently more than 15 phase 3 clinical trials worldwide assessing the efficacy and safety of roxadustat in CKD patients with anemia. This review will summarize recent findings of roxadustat in the treatment of renal anemia. Summary: Although the administration of erythropoiesis-stimulating agents (ESAs) and iron supplementation are a well-established and highly effective therapeutic approach for renal anemia, there are several safety concerns. Current findings from phase 2 and 3 trials suggest that roxadustat is clinically effective and well tolerated. On the one hand, roxadustat could increase endogenous erythropoietin (EPO) levels within or near physiological range in a titratable manner by inducing HIF pathway activation transiently. On the other hand, roxadustat also improves iron metabolism by decreasing serum hepcidin and increasing intestinal iron absorption, which is beneficial to functional iron deficiency and absolute iron deficiency. More importantly, the erythropoietic response of roxadustat is independent of baseline inflammatory state of CKD patients. Thus, the discovery of roxadustat will revolutionize the treatment strategy for renal anemia. Key Messages: Roxadustat is an emerging and promising therapeutic approach against anemia in CKD patients, which differs from those of conventional ESAs. Roxadustat corrects anemia of CKD patients through multiple pathways, beyond elevating EPO levels within physiological range, and also by handling iron metabolism (particularly decreasing the hepcidin levels). Furthermore, the Hb response of roxadustat is independent of the inflammatory microenvironment.

Since the link between uremia and anemia was first described by Richard Bright almost 200 years ago [1], anemia, which contributes to increased morbidity and mortality of patients with chronic kidney disease (CKD) [2], has become one of the most characteristic and visible manifestations of CKD [3]. Despite many advances in understanding of anemia, effective and safe treatment strategies are limited, and a large proportion of CKD patients with anemia still do not meet the hemoglobin (Hb) targets.

Renal anemia is typically characterized as normocytic, normochromic, and hypoproliferative. The data from clinical and preclinical studies suggested the etiology of anemia in CKD is multifactorial, including erythropoietin (EPO) deficiency, abnormal iron metabolism, blood loss, chronic inflammation, reduced erythrocyte survival duration, infection, oxidative stress, and nutritional deficits [4, 5], among which EPO deficiency is most predominant and specific.Findings from various genetic models provide convincing evidence that peritubular interstitial fibroblast-like cells (termed renal EPO-producing cells, RPCs) are the major source of EPO in the kidney [6, 7]. Dysfunction of RPCs, which transdifferentiated into myofibroblasts under conditions of CKD [8], leads to the loss of their ability to synthesize EPO, resulting in EPO deficiency and development of renal anemia. More interestingly, the evidence from genetic fate mapping studies supported the concept that the majority of myofibroblasts were derived from RPCs [9], providing an explanation for correlation with hematocrit, renal EPO mRNA, and renal fibrosis. Moreover, patients who benefited from the recombinant human EPO and related erythropoiesis-stimulating agents (ESAs) [10] also strongly supported the viewpoint that EPO deficiency is the most dominant cause of anemia.

Apart from EPO deficiency, negative iron balance is also a critical contributor to the renal anemia in CKD patients [11, 12]. Increased iron losses (approximately as much as 4–5 g iron per year) [13] and impaired dietary iron absorption (oral iron is no better than placebo and was less effective than intravenous [IV] iron at improving anemia) [14, 15] result in true iron deficiency. In addition, functional iron deficiency, characterized by impaired iron release from body stores, is another key factor for renal anemia [16], manifested by a low transferrin saturation and high ferritin. It should be noted that there is still no effective therapeutic strategy for the management of these patients.

Increasing evidence indicates that hepcidin excess could provide the explanation for disordered iron hemostasis in many CKD patients [17]. It is now well established that hepcidin is the central regulator responsible for maintaining homeostasis of systemic iron. As the sole known exporter responsible for iron export into the plasma, hepcidin could maintain systemic iron balance via regulating the ferroportin, an iron channel on the surface of enterocytes, hepatocytes, macrophages, and placental cells [18]. It is recognized that hepcidin is regulated in response to the signaling pathways about inflammation, iron metabolism, hypoxia, and erythropoietic demand. The best-characterized mechanism is direct transcriptional activation of hepatic hepcidin expression via STAT3 pathway under the condition of inflammatory microenvironment (in particular inflammatory cytokines interleukin-6 [IL-6] and IL-1β) [19]. Indeed, many patients with CKD have a chronic inflammatory state [20]. Recognition of the critical effect of hepcidin on iron metabolism and renal anemia has ignited interest in targeting hepcidin as a new treatment strategy [21].

Currently, the use of ESAs and iron supplementation is the cornerstone of treatment of renal anemia. Although the administration of ESAs and iron supplementation are a well-established and highly effective therapeutic approach for renal anemia, there are several safety concerns. On the one hand, multiple trials of ESAs have demonstrated, at best, no improvement in outcome and, at worst, increased risks of cardiovascular events and mortality [22, 23]. On the other hand, due to the evidence of strong association between labile iron and oxidative stress, bacterial growth, severe gastrointestinal side effects, and hypersensitivity reactions, increased risks of infection and mortality have been concerns related to iron supplementation (in particular IV iron) in CKD patients [24, 25]. As suggested in a recent randomized trial, IV administration of iron is associated with an increased risk of serious adverse events, including cardiovascular and infectious diseases [26]. In fact, detailed concerns have been reviewed [27-29]. Thus, many other approaches have been explored to treat renal anemia [30].

It has been well recognized that rapid induction of EPO production is one of the earliest and most sensitive physiological responses to hypoxia [31]. Actually, EPO synthesized in the kidney, which is the major source of circulating EPO, is regulated by renal oxygen concentration. Until 1992, discovering of hypoxia-inducible factors (HIFs) filled the gap in our understanding of hypoxia leading to erythropoiesis [32]. To date, the molecular mechanisms by which HIF pathway regulates erythropoiesis are well understood. The transcriptional activity of HIF is primarily controlled by its degradation rate, which is regulated by oxygen sensors (known as prolyl-4-hydroxylase domain proteins, PHDs) [33, 34]. In addition to the EPO gene, HIF also targets molecules involved in multiple steps on iron absorption, metabolism, and transport [35]. Although it had been known since the 1980s that EPO transcription and HIF responses were activated with transition metals [36], the discovery of oxygen-dependent PHDs as key regulators of HIF-dependent erythropoiesis provided a theoretical basis for the development of HIF-activating compounds (called HIF-PHIs). Accordingly, recognizing the pivotal roles of the PHD-HIF axis in orchestrating erythropoiesis opened new avenues in the management of renal anemia [37, 38]. The target organs or tissues of HIF-PHIs for anemia correction are summarized in Figure 1.

Fig. 1.

HIF coordinates erythropoietin production with iron metabolism. The oxygen-dependent PHDs as key regulators of HIF-dependent erythropoiesis became the direct therapeutic target for anemia correction. HIF-PHIs stabilize HIF via suppressing the function of PHDs directly. EPO synthesis could be stimulated by HIF (induced by pharmacologic PHD inhibition) in kidney and liver, which plays a central role in stimulating erythropoiesis in the bone marrow. In the duodenum, it is well recognized that DCYTB, DMT1, and FPN are regulated by HIF-2. DCYTB reduces Fe3+ to Fe2+, which then enters enterocytes via DMT1. Iron is then released into the circulation via FPN. The increasing evidence demonstrated that HIF activation could suppress hepcidin expression, which increases FPN expression on enterocytes, hepatocytes, and macrophages, resulting in increased iron absorption and mobilization from internal stores. HRE, hypoxia response element; DCYTB, duodenal cytochrome b reductase 1; DMT1, divalent metal transporter-1; EPO, erythropoietin; EPOR, erythropoietin receptor; RBC, red blood cell; HIF, hypoxia-inducible factor; FPN, ferroportin; Fe2+, ferrous iron; Fe3+, ferric iron.

Fig. 1.

HIF coordinates erythropoietin production with iron metabolism. The oxygen-dependent PHDs as key regulators of HIF-dependent erythropoiesis became the direct therapeutic target for anemia correction. HIF-PHIs stabilize HIF via suppressing the function of PHDs directly. EPO synthesis could be stimulated by HIF (induced by pharmacologic PHD inhibition) in kidney and liver, which plays a central role in stimulating erythropoiesis in the bone marrow. In the duodenum, it is well recognized that DCYTB, DMT1, and FPN are regulated by HIF-2. DCYTB reduces Fe3+ to Fe2+, which then enters enterocytes via DMT1. Iron is then released into the circulation via FPN. The increasing evidence demonstrated that HIF activation could suppress hepcidin expression, which increases FPN expression on enterocytes, hepatocytes, and macrophages, resulting in increased iron absorption and mobilization from internal stores. HRE, hypoxia response element; DCYTB, duodenal cytochrome b reductase 1; DMT1, divalent metal transporter-1; EPO, erythropoietin; EPOR, erythropoietin receptor; RBC, red blood cell; HIF, hypoxia-inducible factor; FPN, ferroportin; Fe2+, ferrous iron; Fe3+, ferric iron.

Close modal

Development History of Roxadustat

Roxadustat was developed almost a decade ago. Derived from its precursor (termed FG-2216), roxadustat (FG-4592), discovered by FibroGen, is a second-generation HIF-PHI, which differs from FG-2216 by the addition of a phenoxy substituent at carbon position seven of the quinoline core [39]. Accordingly, phase 1 clinical trials with roxadustat were initiated in November 2005. Based on the two phase 3 clinical trials in non-dialysis-dependent CKD (NDD-CKD) and dialysis-dependent CKD (DD-CKD) patients, roxadustat received its first approval in China on 17 December 2018 for the treatment of anemia in CKD patients on dialysis (including hemodialysis or peritoneal dialysis) [40]. On 16 August 2019, roxadustat received its approval for the treatment of anemia in NDD-CKD patients in China. Roxadustat has generated an encouraging opportunity for CKD patients by addressing a substantial unmet medical need in the treatment of renal anemia (Fig. 2). Here, we mainly reviewed the results of 3 clinical trials on roxadustat in CKD patients with anemia.

Fig. 2.

Key milestones in the development of roxadustat for the treatment of anemia caused by CKD. CFDA, China Food and Drug Administration; DD-CKD, dialysis-dependent chronic kidney disease; NDA, new drug application; US, United States; NDD-CKD, non-dialysis-dependent chronic kidney disease.

Fig. 2.

Key milestones in the development of roxadustat for the treatment of anemia caused by CKD. CFDA, China Food and Drug Administration; DD-CKD, dialysis-dependent chronic kidney disease; NDA, new drug application; US, United States; NDD-CKD, non-dialysis-dependent chronic kidney disease.

Close modal

Roxadustat in Phase 2 Clinical Trials

The data from six phase 2 clinical trials published so far demonstrated that roxadustat could increase or maintain Hb levels of both non-dialysis and dialysis CKD patients (including ESA-naïve and ESA-converted patients) within the target range with good tolerability [41-46]. And more interestingly, it was demonstrated that roxadustat also had beneficial effects on iron metabolism (significant decreases in serum hepcidin levels particularly). Table 1 summarizes the Hb response and iron parameters in phase 2 clinical studies of roxadustat.

Table 1.

Hb response and iron parameters in phase 2 trials

Hb response and iron parameters in phase 2 trials
Hb response and iron parameters in phase 2 trials

Moreover, roxadustat-induced anemia correction is independent of the degree of baseline inflammation state (assessed by serum C-reactive protein [CRP] levels). Furthermore, in addition to stimulating EPO production and regulating iron homeostasis, roxadustat (including other HIF-PHIs) has been shown to induce nonerythropoietic effects, such as lowering serum low-density lipoprotein or cholesterol levels in CKD patients [46, 47] and blood pressure-lowering effect in clinical or preclinical studies [42, 48]. Detailed results have been reviewed elsewhere [37, 38].

Roxadustat in Phase 3 Clinical Trials

Roxadustat is the first HIF-PHI entering the phase 3 trials. There are currently more than 15 phase 3 clinical trials and a target enrollment of about 10,000 patients worldwide studying the safety, efficacy, and long-term effects of roxadustat in CKD patients, including non-dialysis-dependent, hemodialysis-dependent, peritoneal dialysis-dependent, and patients on newly initiated dialysis [49]. And more importantly, ESA-naïve and ESA-converted patients with various degrees of CKD were also included in those long-term phase 3 studies. Table 2 summarizes the Hb response and iron parameters in phase 3 clinical studies of roxadustat in patients from China.

Table 2.

Hb response and iron parameters in phase 3 trials

Hb response and iron parameters in phase 3 trials
Hb response and iron parameters in phase 3 trials

Notably, two phase 3 trials regarding roxadustat for treatment of renal anemia carried out in China have recently been completed [50, 51]. In an initial 8-week, randomized, double-blind, placebo-controlled phase 3 trial (ClinicalTrials.gov NCT02652819), roxadustat was superior to placebo in increasing Hb levels in CKD patients who were not undergoing dialysis. In this phase 3 trial, the mean ± SD CFB in the Hb level was an increase of 1.9 ± 1.2 g/dL in the roxadustat group and a decrease of 0.4 ± 0.8 g/dL in the placebo group (p < 0.001). Meanwhile, the study showed beneficial effects on iron metabolism (clinically stable serum iron levels, namely no significant difference between the two groups; increased in transferrin levels and total iron-binding capacity; decrease in transferrin saturation and ferritin levels). More interestingly, the reduction of hepcidin level from baseline was 56.14 ± 63.40 and 15.10 ± 48.06 ng/mL, respectively, for a between-group difference of –49.77 ng/mL (95% CI, –66.75 to –32.79).

In another randomized, open-label, active-controlled (epoetin alfa), phase 3 trial (ClinicalTrials.gov NCT02652806), evaluating the noninferiority of roxadustat, which was established if the lower boundary of the two-sided 95% confidence interval for the difference between the values in the roxadustat group and epoetin alfa group was greater than or equal to –1.0 g/dL, in patients undergoing long-term dialysis (at least 16 weeks), the results showed that roxadustat was not inferior to EPO (epoetin alfa) in increasing Hb levels. Although this trial comparing roxadustat with epoetin alfa for 26 weeks showed the noninferiority of roxadustat in the treatment of renal anemia, roxadustat treatment led to a numerically greater mean change in Hb level in CKD patients undergoing dialysis compared with those giving epoetin alfa treatment. Meanwhile, as compared with declined serum iron level in the epoetin alfa group, the serum iron level was clinically stable in the roxadustat group. It should be noted that in this trial, only the use of oral iron therapy was allowed. Actually, during the 26-week treatment period, 32.8% of patients in the roxadustat group received oral iron therapy, as compared with 43% in the epoetin alfa group (p < 0.001). Despite increased transferrin level and total iron-binding capacity, the decreased transferrin saturation supports the absorption-promoting effect on enteric iron with roxadustat. On the other hand, consistent with the results of previous phase 2 trials, hepcidin decreased significantly with roxadustat. Furthermore, the phase 3 trial of roxadustat involving patients undergoing hemodialysis addressed one important question, namely, the efficacy of roxadustat for CKD patients with an apparent inflammatory state (as assessed on the basis of CRP levels). Interestingly, among patients with elevated CRP levels, a greater increase in the Hb level response with roxadustat than those responses with epoetin alfa (despite receiving higher doses of epoetin alfa) was observed. Therefore, consistent with results in phase 2 studies of roxadustat, the results of phase 3 trials demonstrated that roxadustat could also improve anemia in CKD patients with inflammation. However, the detailed mechanisms are still not available. It is currently recognized that increased hepcidin expression in the liver, which leads to iron deficiency, could be induced by inflammation. Thus, the hepcidin-lowering effect of roxadustat could contribute to these findings in CKD patients with inflammatory state.

Recently, another phase 3 randomized, open-label, 24-week study (ClinicalTrials.gov NCT02780726) investigating the efficacy and safety of roxadustat in anemic Japanese patients on peritoneal dialysis who previously received ESA (ESA-converted group) or not (ESA-naïve group) has also been published [52]. Consistent with the results from the above two trials, roxadustat was effective in maintaining target Hb levels (92.3% in the ESA-naïve group and 74.4% in the ESA-converted group) in anemic patients on peritoneal dialysis. Meanwhile, the results also showed that iron metabolism parameters remained clinically stable throughout the study (only 1 patient was treated with IV iron), suggesting an increase in iron bioavailability via improving iron mobilization and absorption mediated by the observed reduction in serum hepcidin levels from baseline (also supported by the increase in transferrin). More importantly, roxadustat was generally safe and well tolerated in this study.

Roxadustat is an orally bioavailable, reversible HIF-PHI that mimics a novel way of utilizing the body’s natural compensatory mechanisms in response to hypoxia, promoting HIF transcriptional activity. The data from trials illustrated that roxadustat corrects anemia of CKD patients with some distinct advantages.

One of the most striking superiorities is that roxadustat could induce a transient activation of HIF and increase in expression of HIF-regulated genes. With a half-life of approximately 12 h, roxadustat, administered twice or thrice weekly, enables HIF transcriptional activity to return to baseline between doses, which results in induction of expression of EPO in a titratable manner [53]. Furthermore, roxadustat transiently increased endogenous EPO levels within or near physiological range in patients with anemia of CKD. Thus, it appears that anemia correction with roxadustat could avoid these potential adverse effects caused by high ESA doses.

Beyond elevating EPO level, roxadustat could correct anemia by handling iron metabolism and particularly by decreasing the hepcidin levels, a response that was independent of baseline CRP levels and disease states of CKD patients (not receiving dialysis, hemodialysis, or peritoneal dialysis). This allows access to improve functional iron deficiency as well as increased absorption of oral iron, thus avoiding the use of high doses of IV iron, which has been associated with an increased inflammatory state and increased mortality [25]. The increase in transferrin induced by roxadustat is a direct effect of HIF activation, as there are two HIF binding sites in the 5′ enhancer region of the gene encoding transferrin [54]. However, it remains unclear how roxadustat is responsible for hepcidin suppression.

CKD anemia is also a type of chronic inflammatory anemia, which influences the erythropoiesis processes via inhibition of EPO production and disturbing bone marrow erythroblastic development and iron metabolism [55]. The data from phase 2 and 3 trials demonstrated that the erythropoietic response of roxadustat appears to be independent of inflammation, as correction and maintenance dose requirements were not associated with baseline CRP levels. This independence of response from the inflammatory state of patients implies that roxadustat might be one of the appropriate clinical strategies to treat anemia of CKD patients who have a hyporesponsiveness to ESA due to the high inflammatory state.

In addition, the decreased total cholesterol level was greater with roxadustat than with the control in phase 2 and 3 clinical trials. Meanwhile, compared with the epoetin alfa treatment, hypertension occurred at a lower frequency in patients with roxadustat, which has proved to be beneficial to cardiovascular events. The possible advantages of roxadustat are summarized in Table 3.

Table 3.

Potential advantages of roxadustat based on clinical trials

Potential advantages of roxadustat based on clinical trials
Potential advantages of roxadustat based on clinical trials

Although the data from phase 2 and 3 clinical trials in patients with CKD with or without dialysis have clearly demonstrated that roxadustat is effective and well tolerated, several important questions relating to the safety concerns need to be answered [56].

Accruing evidence has shown that HIF transcription factor directly regulates hundreds of genes, and consequently plays an important role in a broad spectrum of cellular functions and biological processes other than erythropoiesis, including energy metabolism, angiogenesis, mitochondrial metabolism, cellular growth and differentiation, inflammation, cell motility, matrix production, and epigenetics [57-59]. Therefore, it is reasonable to speculate that HIF stabilization by pharmacologic PHD inhibition will have a range of nonerythropoietic effects. Both phase 3 trials have shown a slightly higher risk of hyperkalemia with roxadustat than with control (epoietin alfa or placebo). In addition, the incidence of metabolic acidosis that was reported as adverse events was higher in the roxadustat group than in the control group (placebo) [51]. Furthermore, a potential proangiogenic effect related to the vascular endothelial growth factor (VEGF) and VEGF receptors, the possibility of development of pulmonary hypertension, and the potential progression of kidney disease related to the HIF activation over long periods are three intriguing questions [60, 61]. Accordingly, it is reasonable to await in-progress trials (ClinicalTrials.gov numbers NCT02052310, NCT02273726, and NCT01750190) and to proceed with additional studies reexamining the risks and benefits of normalization of the Hb level in patients with anemia of CKD and carefully monitoring for as yet unappreciated problems with roxadustat.

In summary, reduced EPO production and disorder of iron metabolism are two main characteristics of renal anemia. Roxadustat, as the first HIF-PHI, has been proven to exert an erythropoietic role in renal anemia just like one stone kills two birds by elevating the physiological range of endogenous EPO production and reducing hepcidin. In addition, roxadustat has been shown to successfully increase Hb level even with an inflammatory microenvironment, a status of CKD patients refractory to the current treatment of ESAs. It is thereby recognized that roxadustat could serve as a unique promising therapeutic approach against renal anemia differing from conventional ESAs. Although the data from phase 2 and phase 3 clinical trials indicate that roxadustat is an effective alternative for renal anemia with good tolerability, it should be noted that more large-scale, long-term clinical trials for roxadustat in the treatment of renal anemia are still needed for a better understanding of its safety and nonerythropoietic effects.

The authors have no conflicts of interest to declare.

This study was supported by grants from the National Natural Scientific Foundation (No. 81720108007 and 81670696), the National Key Research and Development Program (No. 2018YFC1314000), and the Clinic Research Center of Jiangsu Province (No. BL2014080).

Z.-L. Li and Y. Tu wrote the manuscript. B.-C. Liu supervised the work and revised the manuscript.

1.
Bright
R
.
Cases and Observations Illustrative of Renal Disease, Accompanied with the Secretion of Albuminous Urine
.
Med Chir Rev
.
1836
Jul
;
25
(
49
):
23
35
.
[PubMed]
2.
Hörl
WH
.
Anaemia management and mortality risk in chronic kidney disease
.
Nat Rev Nephrol
.
2013
May
;
9
(
5
):
291
301
.
[PubMed]
1759-5061
3.
Stauffer
ME
,
Fan
T
.
Prevalence of anemia in chronic kidney disease in the United States
.
PLoS One
.
2014
Jan
;
9
(
1
):
e84943
.
[PubMed]
1932-6203
4.
Babitt
JL
,
Lin
HY
.
Mechanisms of anemia in CKD
.
J Am Soc Nephrol
.
2012
Oct
;
23
(
10
):
1631
4
.
[PubMed]
1046-6673
5.
Fishbane
S
,
Spinowitz
B
.
Update on Anemia in ESRD and Earlier Stages of CKD: core Curriculum 2018
.
Am J Kidney Dis
.
2018
Mar
;
71
(
3
):
423
35
.
[PubMed]
0272-6386
6.
Maxwell
PH
,
Osmond
MK
,
Pugh
CW
,
Heryet
A
,
Nicholls
LG
,
Tan
CC
, et al.
Identification of the renal erythropoietin-producing cells using transgenic mice
.
Kidney Int
.
1993
Nov
;
44
(
5
):
1149
62
.
[PubMed]
0085-2538
7.
Semenza
GL
,
Koury
ST
,
Nejfelt
MK
,
Gearhart
JD
,
Antonarakis
SE
.
Cell-type-specific and hypoxia-inducible expression of the human erythropoietin gene in transgenic mice
.
Proc Natl Acad Sci USA
.
1991
Oct
;
88
(
19
):
8725
9
.
[PubMed]
0027-8424
8.
Souma
T
,
Yamazaki
S
,
Moriguchi
T
,
Suzuki
N
,
Hirano
I
,
Pan
X
, et al.
Plasticity of renal erythropoietin-producing cells governs fibrosis
.
J Am Soc Nephrol
.
2013
Oct
;
24
(
10
):
1599
616
.
[PubMed]
1046-6673
9.
Falke
LL
,
Gholizadeh
S
,
Goldschmeding
R
,
Kok
RJ
,
Nguyen
TQ
.
Diverse origins of the myofibroblast—implications for kidney fibrosis
.
Nat Rev Nephrol
.
2015
Apr
;
11
(
4
):
233
44
.
[PubMed]
1759-5061
10.
Robles
NR
.
The Safety of Erythropoiesis-Stimulating Agents for the Treatment of Anemia Resulting from Chronic Kidney Disease
.
Clin Drug Investig
.
2016
Jun
;
36
(
6
):
421
31
.
[PubMed]
1173-2563
11.
Lopez
A
,
Cacoub
P
,
Macdougall
IC
,
Peyrin-Biroulet
L
.
Iron deficiency anaemia
.
Lancet
.
2016
Feb
;
387
(
10021
):
907
16
.
[PubMed]
0140-6736
12.
Panwar
B
,
Gutiérrez
OM
.
Disorders of Iron Metabolism and Anemia in Chronic Kidney Disease
.
Semin Nephrol
.
2016
Jul
;
36
(
4
):
252
61
.
[PubMed]
0270-9295
13.
Macdougall
IC
,
Bircher
AJ
,
Eckardt
KU
,
Obrador
GT
,
Pollock
CA
,
Stenvinkel
P
, et al.;
Conference Participants
.
Iron management in chronic kidney disease: conclusions from a “Kidney Disease: Improving Global Outcomes” (KDIGO) Controversies Conference
.
Kidney Int
.
2016
Jan
;
89
(
1
):
28
39
.
[PubMed]
0085-2538
14.
Markowitz
GS
,
Kahn
GA
,
Feingold
RE
,
Coco
M
,
Lynn
RI
.
An evaluation of the effectiveness of oral iron therapy in hemodialysis patients receiving recombinant human erythropoietin
.
Clin Nephrol
.
1997
Jul
;
48
(
1
):
34
40
.
[PubMed]
0301-0430
15.
Macdougall
IC
,
Tucker
B
,
Thompson
J
,
Tomson
CR
,
Baker
LR
,
Raine
AE
.
A randomized controlled study of iron supplementation in patients treated with erythropoietin
.
Kidney Int
.
1996
Nov
;
50
(
5
):
1694
9
.
[PubMed]
0085-2538
16.
Gafter-Gvili
A
,
Schechter
A
,
Rozen-Zvi
B
.
Iron Deficiency Anemia in Chronic Kidney Disease
.
Acta Haematol
.
2019
;
142
(
1
):
44
50
.
[PubMed]
0001-5792
17.
Ueda
N
,
Takasawa
K
.
Role of Hepcidin-25 in Chronic Kidney Disease: anemia and Beyond
.
Curr Med Chem
.
2017
;
24
(
14
):
1417
52
.
[PubMed]
0929-8673
18.
Nemeth
E
,
Tuttle
MS
,
Powelson
J
,
Vaughn
MB
,
Donovan
A
,
Ward
DM
, et al.
Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization
.
Science
.
2004
Dec
;
306
(
5704
):
2090
3
.
[PubMed]
0036-8075
19.
Pietrangelo
A
,
Dierssen
U
,
Valli
L
,
Garuti
C
,
Rump
A
,
Corradini
E
, et al.
STAT3 is required for IL-6-gp130-dependent activation of hepcidin in vivo
.
Gastroenterology
.
2007
Jan
;
132
(
1
):
294
300
.
[PubMed]
0016-5085
20.
Akchurin
OM
,
Kaskel
F
.
Update on inflammation in chronic kidney disease
.
Blood Purif
.
2015
;
39
(
1-3
):
84
92
.
[PubMed]
0253-5068
21.
Malyszko
J
,
Malyszko
JS
,
Matuszkiewicz-Rowinska
J
.
Hepcidin as a therapeutic target for anemia and inflammation associated with chronic kidney disease
.
Expert Opin Ther Targets
.
2019
May
;
23
(
5
):
407
21
.
[PubMed]
1472-8222
22.
Solomon
SD
,
Uno
H
,
Lewis
EF
,
Eckardt
KU
,
Lin
J
,
Burdmann
EA
, et al.;
Trial to Reduce Cardiovascular Events with Aranesp Therapy (TREAT) Investigators
.
Erythropoietic response and outcomes in kidney disease and type 2 diabetes
.
N Engl J Med
.
2010
Sep
;
363
(
12
):
1146
55
.
[PubMed]
0028-4793
23.
Koulouridis
I
,
Alfayez
M
,
Trikalinos
TA
,
Balk
EM
,
Jaber
BL
.
Dose of erythropoiesis-stimulating agents and adverse outcomes in CKD: a metaregression analysis
.
Am J Kidney Dis
.
2013
Jan
;
61
(
1
):
44
56
.
[PubMed]
0272-6386
24.
Li
X
,
Cole
SR
,
Kshirsagar
AV
,
Fine
JP
,
Stürmer
T
,
Brookhart
MA
.
Safety of Dynamic Intravenous Iron Administration Strategies in Hemodialysis Patients
.
Clin J Am Soc Nephrol
.
2019
May
;
14
(
5
):
728
37
.
[PubMed]
1555-9041
25.
Bailie
GR
,
Larkina
M
,
Goodkin
DA
,
Li
Y
,
Pisoni
RL
,
Bieber
B
, et al.
Data from the Dialysis Outcomes and Practice Patterns Study validate an association between high intravenous iron doses and mortality
.
Kidney Int
.
2015
Jan
;
87
(
1
):
162
8
.
[PubMed]
0085-2538
26.
Agarwal
R
,
Kusek
JW
,
Pappas
MK
.
A randomized trial of intravenous and oral iron in chronic kidney disease
.
Kidney Int
.
2015
Oct
;
88
(
4
):
905
14
.
[PubMed]
0085-2538
27.
Del Vecchio
L
,
Locatelli
F
.
An overview on safety issues related to erythropoiesis-stimulating agents for the treatment of anaemia in patients with chronic kidney disease
.
Expert Opin Drug Saf
.
2016
Aug
;
15
(
8
):
1021
30
.
[PubMed]
1474-0338
28.
Slotki
I
,
Cabantchik
ZI
.
The Labile Side of Iron Supplementation in CKD
.
J Am Soc Nephrol
.
2015
Nov
;
26
(
11
):
2612
9
.
[PubMed]
1046-6673
29.
Li
X
,
Kshirsagar
AV
,
Brookhart
MA
.
Safety of intravenous iron in hemodialysis patients
.
Hemodial Int
.
2017
Jun
;
21
Suppl 1
:
S93
103
.
[PubMed]
1492-7535
30.
Bonomini
M
,
Del Vecchio
L
,
Sirolli
V
,
Locatelli
F
.
New Treatment Approaches for the Anemia of CKD
.
Am J Kidney Dis
.
2016
Jan
;
67
(
1
):
133
42
.
[PubMed]
0272-6386
31.
Luks
AM
. Physiology in Medicine: A physiologic approach to prevention and treatment of acute high-altitude illnesses. J Appl Physiol (
1985
). 2015 Mar;118(5):509-19.
32.
Semenza
GL
,
Wang
GL
.
A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation
.
Mol Cell Biol
.
1992
Dec
;
12
(
12
):
5447
54
.
[PubMed]
0270-7306
33.
Jaakkola
P
,
Mole
DR
,
Tian
YM
,
Wilson
MI
,
Gielbert
J
,
Gaskell
SJ
, et al.
Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation
.
Science
.
2001
Apr
;
292
(
5516
):
468
72
.
[PubMed]
0036-8075
34.
Kaelin
WG
 Jr
,
Ratcliffe
PJ
.
Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway
.
Mol Cell
.
2008
May
;
30
(
4
):
393
402
.
[PubMed]
1097-2765
35.
Koury
MJ
,
Haase
VH
.
Anaemia in kidney disease: harnessing hypoxia responses for therapy
.
Nat Rev Nephrol
.
2015
Jul
;
11
(
7
):
394
410
.
[PubMed]
1759-5061
36.
Edwards
MS
,
Curtis
JR
.
Use of cobaltous chloride in anaemia of maintenance hemodialysis patients
.
Lancet
.
1971
Sep
;
2
(
7724
):
582
3
.
[PubMed]
0140-6736
37.
Gupta
N
,
Wish
JB
.
Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitors: A Potential New Treatment for Anemia in Patients With CKD
.
Am J Kidney Dis
.
2017
Jun
;
69
(
6
):
815
26
.
[PubMed]
0272-6386
38.
Joharapurkar
AA
,
Pandya
VB
,
Patel
VJ
,
Desai
RC
,
Jain
MR
.
Prolyl Hydroxylase Inhibitors: A Breakthrough in the Therapy of Anemia Associated with Chronic Diseases
.
J Med Chem
.
2018
Aug
;
61
(
16
):
6964
82
.
[PubMed]
0022-2623
39.
Rabinowitz
MH
.
Inhibition of hypoxia-inducible factor prolyl hydroxylase domain oxygen sensors: tricking the body into mounting orchestrated survival and repair responses
.
J Med Chem
.
2013
Dec
;
56
(
23
):
9369
402
.
[PubMed]
0022-2623
40.
FibroGen
. FibroGen Announces Approval of Roxadustat in China for the Treatment of Anemia in Chronic Kidney Disease Patients on Dialysis. [Media release]. Dec
2018
. http://investor.fibrogen.com/news-releases/news-release-details/fibrogen-announces-approval-roxadustat-china-treatment-anemia
41.
Besarab
A
,
Provenzano
R
,
Hertel
J
,
Zabaneh
R
,
Klaus
SJ
,
Lee
T
, et al.
Randomized placebo-controlled dose-ranging and pharmacodynamics study of roxadustat (FG-4592) to treat anemia in nondialysis-dependent chronic kidney disease (NDD-CKD) patients
.
Nephrol Dial Transplant
.
2015
Oct
;
30
(
10
):
1665
73
.
[PubMed]
0931-0509
42.
Besarab
A
,
Chernyavskaya
E
,
Motylev
I
,
Shutov
E
,
Kumbar
LM
,
Gurevich
K
, et al.
Roxadustat (FG-4592): Correction of Anemia in Incident Dialysis Patients
.
J Am Soc Nephrol
.
2016
Apr
;
27
(
4
):
1225
33
.
[PubMed]
1046-6673
43.
Provenzano
R
,
Besarab
A
,
Sun
CH
,
Diamond
SA
,
Durham
JH
,
Cangiano
JL
, et al.
Oral Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor Roxadustat (FG-4592) for the Treatment of Anemia in Patients with CKD
.
Clin J Am Soc Nephrol
.
2016
Jun
;
11
(
6
):
982
91
.
[PubMed]
1555-9041
44.
Provenzano
R
,
Besarab
A
,
Wright
S
,
Dua
S
,
Zeig
S
,
Nguyen
P
, et al.
Roxadustat (FG-4592) Versus Epoetin Alfa for Anemia in Patients Receiving Maintenance Hemodialysis: A Phase 2, Randomized, 6- to 19-Week, Open-Label, Active-Comparator, Dose-Ranging, Safety and Exploratory Efficacy Study
.
Am J Kidney Dis
.
2016
Jun
;
67
(
6
):
912
24
.
[PubMed]
0272-6386
45.
Chen
N
,
Qian
J
,
Chen
J
,
Yu
X
,
Mei
C
,
Hao
C
, et al.
Phase 2 studies of oral hypoxia-inducible factor prolyl hydroxylase inhibitor FG-4592 for treatment of anemia in China
.
Nephrol Dial Transplant
.
2017
Aug
;
32
(
8
):
1373
86
.
[PubMed]
0931-0509
46.
Akizawa
T
,
Iwasaki
M
,
Otsuka
T
,
Reusch
M
,
Misumi
T
.
Roxadustat Treatment of Chronic Kidney Disease-Associated Anemia in Japanese Patients Not on Dialysis: A Phase 2, Randomized, Double-Blind, Placebo-Controlled Trial
.
Adv Ther
.
2019
Jun
;
36
(
6
):
1438
54
.
[PubMed]
0741-238X
47.
Parmar
DV
,
Kansagra
KA
,
Patel
JC
,
Joshi
SN
,
Sharma
NS
,
Shelat
AD
, et al.;
on behalf of the ZYAN1 Trial Investigators
.
Outcomes of Desidustat Treatment in People with Anemia and Chronic Kidney Disease: A Phase 2 Study
.
Am J Nephrol
.
2019
;
49
(
6
):
470
8
.
[PubMed]
0250-8095
48.
Flamme
I
,
Oehme
F
,
Ellinghaus
P
,
Jeske
M
,
Keldenich
J
,
Thuss
U
.
Mimicking hypoxia to treat anemia: HIF-stabilizer BAY 85-3934 (Molidustat) stimulates erythropoietin production without hypertensive effects
.
PLoS One
.
2014
Nov
;
9
(
11
):
e111838
.
[PubMed]
1932-6203
49.
Dhillon
S
.
Roxadustat: First Global Approval
.
Drugs
.
2019
Apr
;
79
(
5
):
563
72
.
[PubMed]
0012-6667
50.
Chen
N
,
Hao
C
,
Liu
BC
,
Lin
H
,
Wang
C
,
Xing
C
, et al.
Roxadustat Treatment for Anemia in Patients Undergoing Long-Term Dialysis
.
N Engl J Med
.
2019
Sep
;
381
(
11
):
1011
22
.
[PubMed]
0028-4793
51.
Chen
N
,
Hao
C
,
Peng
X
,
Lin
H
,
Yin
A
,
Hao
L
, et al.
Roxadustat for Anemia in Patients with Kidney Disease Not Receiving Dialysis
.
N Engl J Med
.
2019
Sep
;
381
(
11
):
1001
10
.
[PubMed]
0028-4793
52.
Akizawa
T
,
Otsuka
T
,
Reusch
M
,
Ueno
M
.
Intermittent Oral Dosing of Roxadustat in Peritoneal Dialysis Chronic Kidney Disease Patients with Anemia: A Randomized, Phase 3, Multicenter, Open-Label Study
.
Ther Apher Dial
.
2019
Jun
;
•••
:
1744-9987.12888
.
[PubMed]
1744-9979
53.
Bernhardt
WM
,
Wiesener
MS
,
Scigalla
P
,
Chou
J
,
Schmieder
RE
,
Günzler
V
, et al.
Inhibition of prolyl hydroxylases increases erythropoietin production in ESRD
.
J Am Soc Nephrol
.
2010
Dec
;
21
(
12
):
2151
6
.
[PubMed]
1046-6673
54.
Rolfs
A
,
Kvietikova
I
,
Gassmann
M
,
Wenger
RH
.
Oxygen-regulated transferrin expression is mediated by hypoxia-inducible factor-1
.
J Biol Chem
.
1997
Aug
;
272
(
32
):
20055
62
.
[PubMed]
0021-9258
55.
Yilmaz
MI
,
Solak
Y
,
Covic
A
,
Goldsmith
D
,
Kanbay
M
.
Renal anemia of inflammation: the name is self-explanatory
.
Blood Purif
.
2011
;
32
(
3
):
220
5
.
[PubMed]
0253-5068
56.
Kaplan
J
.
Roxadustat and Anemia of Chronic Kidney Disease
.
N Engl J Med
.
2019
Sep
;
381
(
11
):
1070
2
.
[PubMed]
0028-4793
57.
Semenza
GL
.
Oxygen sensing, hypoxia-inducible factors, and disease pathophysiology
.
Annu Rev Pathol
.
2014
;
9
(
1
):
47
71
.
[PubMed]
1553-4006
58.
Choudhry
H
,
Harris
AL
.
Advances in Hypoxia-Inducible Factor Biology
.
Cell Metab
.
2018
Feb
;
27
(
2
):
281
98
.
[PubMed]
1550-4131
59.
Li
ZL
,
Lv
LL
,
Tang
TT
,
Wang
B
,
Feng
Y
,
Zhou
LT
, et al.
HIF-1α inducing exosomal microRNA-23a expression mediates the cross-talk between tubular epithelial cells and macrophages in tubulointerstitial inflammation
.
Kidney Int
.
2019
Feb
;
95
(
2
):
388
404
.
[PubMed]
0085-2538
60.
Maxwell
PH
,
Eckardt
KU
.
HIF prolyl hydroxylase inhibitors for the treatment of renal anaemia and beyond
.
Nat Rev Nephrol
.
2016
Mar
;
12
(
3
):
157
68
.
[PubMed]
1759-5061
61.
Li
ZL
,
Lv
LL
,
Wang
B
,
Tang
TT
,
Feng
Y
,
Cao
JY
, et al.
The profibrotic effects of MK-8617 on tubulointerstitial fibrosis mediated by the KLF5 regulating pathway. FASEB J.
2019
Aug 26:fj201901087RR.
Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (CC BY-NC-ND). Usage and distribution for commercial purposes as well as any distribution of modified material requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.