Skip to main content
Erschienen in: Advances in Therapy 1/2021

Open Access 29.10.2020 | Review

Burden of Anemia in Chronic Kidney Disease: Beyond Erythropoietin

verfasst von: Ramy M. Hanna, Elani Streja, Kamyar Kalantar-Zadeh

Erschienen in: Advances in Therapy | Ausgabe 1/2021

Abstract

Anemia is a frequent comorbidity of chronic kidney disease (CKD) and is associated with a considerable burden because of decreased patient health-related quality of life and increased healthcare resource utilization. Based on observational data, anemia is associated with an increased risk of CKD progression, cardiovascular events, and all-cause mortality. The current standard of care includes oral or intravenous iron supplementation, erythropoiesis-stimulating agents, and red blood cell transfusion. However, each of these therapies has its own set of population-specific patient concerns, including increased risk of cardiovascular disease, thrombosis, and mortality. Patients receiving dialysis or those who have concurrent diabetes or high blood pressure may be at greater risk of developing these complications. In particular, treatment with high doses of erythropoiesis-stimulating agents has been associated with increased rates of hospitalization, cardiovascular events, and mortality. Resistance to erythropoiesis-stimulating agents remains a therapeutic challenge in a subset of patients. Hypoxia-inducible factor transcription factors, which regulate several genes involved in erythropoiesis and iron metabolism, can be stabilized by a new class of drugs that act as inhibitors of hypoxia-inducible factor prolyl-hydroxylase enzymes to promote erythropoiesis and elevate hemoglobin levels. Here, we review the burden of anemia of chronic kidney disease, the shortcomings of current standard of care, and the potential practical advantages of hypoxia-inducible factor prolyl-hydroxylase inhibitors in the treatment of patients with anemia of CKD.
Key Summary Points
Anemia is common in patients with chronic kidney disease and has been associated with increased risk of cardiovascular morbidity and mortality in observational studies as well as decreased patient quality of life and increased healthcare utilization.
The current standard of care includes supplemental iron, erythropoiesis-stimulating agents, and red blood cell transfusions, although each has drawbacks.
High doses of erythropoiesis-stimulating agents have been associated with increased cardiovascular complications and mortality.
Hypoxia-inducible factor-prolyl hydroxylase inhibitors are novel treatments for anemia of chronic kidney disease that prevent degradation of the transcription factor hypoxia-inducible factor, which stimulates erythropoiesis to physiologic levels.

Digital Features

This article is published with digital features, including a summary slide, to facilitate understanding of the article. To view digital features for this article go to https://​doi.​org/​10.​6084/​m9.​figshare.​13035146.

Introduction

Anemia is a common complication of chronic kidney disease (CKD), representing a significant burden to patients and healthcare systems [1, 2]. According to the Kidney Disease: Improving Global Outcomes (KDIGO) clinical practice guidelines, anemia of CKD is defined as hemoglobin (Hb) < 13.0 g/dl for men and < 12.0 g/dl for nonpregnant women [3] and largely results from decreased erythropoietin (EPO) production by the failing kidney and/or altered iron homeostasis [4, 5]. The current standard of care for anemia of CKD includes oral or intravenous (IV) iron, erythropoiesis-stimulating agents (ESAs), and red blood cell (RBC) transfusion, each of which has potential problems and variable effectiveness [2, 3]. The impact of anemia correction on patient health-related quality of life (HR-QOL) is unknown, and persistent safety issues contribute to uncertainty regarding the optimal target Hb. This article reviews the burden of anemia of CKD, including its impact on mortality and cardiovascular risk, HR-QOL, hospitalization and transfusion needs, iron supplementation needs, the conservative management of CKD to delay dialysis, end-stage renal disease (ESRD) transition outcomes, anemia management at home, and anemia management in transplant recipients. An assessment of the risk to the benefit profile associated with current standard of care and discussion surrounding novel agents in development based on alternative erythropoietic mechanisms are also provided. This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.

Disease Burden

Prevalence

The estimated global prevalence of CKD is 11% for patients with CKD stage 3 [estimated glomerular filtration rate (eGFR) < 60 ml/min/1.73 m2] to stage 5 (eGFR < 15 ml/min/1.73 m2) and 13% for patients with CKD stage 1 (albumin-to-creatine ratio > 30 plus eGFR > 90 ml/min/1.73 m2) to stage 5 [6]. In the US, the prevalence of stage 1–5 CKD was 14.0% (representing ~ 31.4 million people) according to the 2007–2010 data from the National Health and Nutrition Examination Survey (NHANES) [7]. Similarly, the US Centers for Disease Control and Prevention estimated that the prevalence of CKD stage 1 to stage 4 (eGFR 15–29 ml/min/1.73 m2) was 15% (~37 million people) in 2013–2016 [8].
Anemia prevalence increases with CKD stage. In the NHANES analysis, 15.4% (~4.8 million people) had anemia of CKD, and anemia prevalence was 17.4%, 50.3%, and 53.4% in stages 3, 4, and 5 CKD, respectively [7]. Anemia of CKD prevalence also increases in patients with comorbidities and with age, from 28.0% in those aged 18–63 years to 50.1% in those aged ≥ 66 years among US patients with non-dialysis-dependent (NDD) CKD [1].

Cardiovascular Risk and Mortality

Anemia, fluid overload, and arteriovenous fistulas can lead to volume overload that ultimately results in cardiomyopathy, including increased left ventricular hypertrophy (LVH), and systolic and diastolic dysfunction [9, 10]. This cardiomyopathy may present as ischemic heart disease or heart failure, even when arterial vascular disease is absent [10]. Anemia has been associated with an increased risk of cardiovascular events and all-cause mortality in a number of observational studies [1118], and the American Heart Association considers anemia to be a nontraditional (non-Framingham) cardiovascular risk factor in patients with CKD [10]. In a US study of > 900,000 patients with NDD-CKD, functional iron deficiency anemia was associated with an increased risk of mortality [hazard ratio (HR) 1.11, 95% CI 1.07–1.14] and an increased relative risk (RR) of cardiovascular hospitalization after 1 year (RR 1.21, 95% CI 1.12–1.30) and 2 years (RR 1.13, 95% CI 1.07–1.21) [11]. Similarly, a Danish study of patients with dialysis-dependent-CKD (DD-CKD) and NDD-CKD found that anemia was associated with increased risks of major adverse cardiovascular events (MACE), acute hospitalization, and all-cause death [12], and a Japanese study of NDD-CKD patients reported that isolated anemia and iron deficiency anemia were associated with increased risks of cardiovascular-related and all-cause mortality [13]. After adjusting for other cardiovascular risk factors (including age, diabetes, hypertension, and dyslipidemia), patients with anemia in the US Atherosclerosis Risk in Communities (ARIC) study had a significantly increased risk of stroke with comorbid CKD versus no CKD (HR 5.43, 95% CI 2.04–14.41), whereas in patients without anemia, the risk of stroke with CKD was not significantly increased (HR 1.41, 95% CI 0.93–2.14) [14]. In patients with diabetes, a pooled analysis of data from the ARIC, Cardiovascular Health, Framingham Heart, and Framingham Offspring studies found an association between anemia and increased risks of the individual and composite outcomes of myocardial infarction (MI), fatal coronary heart disease, stroke, or death, and all-cause mortality among patients with comorbid CKD, but not in those without CKD [15]. An association between low Hb levels and increased risks of cardiovascular and all-cause mortality was also observed in a Korean study of ~300,000 patients without cardiovascular disease [16]. Furthermore, anemia was associated with increased cardiovascular risk among Japanese patients undergoing treatment for hypertension [17] and in an Italian study of patients with diabetes [18].
However, the association between anemia and cardiovascular morbidity and mortality in patients with CKD is primarily based on observational studies, and randomized interventional trials have yet to demonstrate a reduction in mortality risk with correction of anemia [19]. Notably, clinical trials that attempted to raise Hb to high levels (13–13.5 g/dl) with darbepoetin alfa therapy found an increased risk of mortality or cardiovascular- or renal-related complications compared with a near-normal or low Hb target (11.3 g/dl; HR 1.34, 95% CI 1.03–1.74, P = 0.03) [20] and also an increased risk of fatal or non-fatal stroke compared with placebo (HR 1.92, 95% CI 1.38–2.68, P < 0.001) [21].
Anemia of CKD represents an independent risk factor for poor HR-QOL [22]. In patients with CKD anemia, cardiovascular complications are associated with significantly impaired HR-QOL (EQ-5D visual analog scale coefficient −5.68, P = 0.028) and work productivity (Work Productivity and Activity Impairment questionnaire: activity impairment coefficient 8.04, P = 0.032) compared with non-anemic CKD patients [23]. The Centers for Medicare and Medicaid Services states that all dialysis units should actively monitor patient HR-QOL, underscoring the need to understand long-term HR-QOL implications when treating anemia and other comorbidities in patients with CKD [24].

Healthcare Resource Use

The high prevalence of anemia of CKD represents an important clinical and economic healthcare burden [25]. Patients with moderate CKD and severe anemia (Hb ≤ 9 g/dl) generally require increased hospitalization compared with those without severe anemia [26]. Because patients with CKD and anemia use more overall healthcare resources, their care incurs more costs than those without anemia [1]. In the US, patients with anemia of CKD have estimated total healthcare costs of US$3800–US$4800/patient-month [27]; yearly treatment costs among US patients with CKD are estimated to be more than three-fold higher in patients with anemia than in those without anemia [28].

Current Standard of Care

Current treatment options for anemia include oral or IV iron, ESAs, and RBC transfusion (Table 1). Although raising Hb levels can lead to improved HR-QOL, morbidity, mortality, and reduced hospitalization [29, 30], increasing Hb to “normal” levels has led to adverse outcomes highlighting the issues associated with the current standard of care for anemia of CKD.
Table 1
Pros and cons of pharmacologic treatment for anemia of chronic kidney disease
 
Short-acting ESAs
Long-acting ESAs
HIF-PH inhibitors
Pros
Reduces need for RBC transfusions [31]
May reduce fatigue and improve HR-QOL [29]
IV administration is preferred in patients on hemodialysis [32]
Reduces need for RBC transfusions [21]
May reduce fatigue and improve HR-QOL [29]
Can be administered less frequently than short-acting ESAs [33]
May be cheaper than short-acting ESAs [34]
IV administration is preferred in patients on hemodialysis [32]
Have been shown to be noninferior to ESAs in raising or maintaining Hb [35]
Can be administered orally [36]
May reduce the need for iron supplementation by mobilizing stored iron [37]
Cons
Higher doses required to reach high Hb targets may increase risk of adverse cardiovascular outcomes [20]
Often requires supplemental iron administration [3]
Administered 3 times per week [31]
Higher doses required to reach high Hb targets may increase risk of adverse cardiovascular outcomes [21]
Often requires supplemental iron administration [3]
May confer increased risk of mortality compared with short-acting ESAs [38]
Additional research needed to evaluate potential effects on tumor growth [36]
ESA erythropoiesis-stimulating agent, Hb hemoglobin, HIF-PH hypoxia-inducible factor prolyl-hydroxylase, HR-QOL health-related quality of life, IV intravenous, RBC red blood cell

Iron

Iron deficiency frequently presents in patients with CKD and is mediated by hepcidin, a hepatic peptide that inhibits iron absorption and release from iron stores and macrophages [5]. Iron deficiency is compounded by increased iron demands with ESAs, which can limit their effectiveness [39]. Supplementary iron can improve physical, cognitive, and immune function [40]. Although less expensive and safer than IV iron, oral iron is poorly absorbed and associated with gastrointestinal adverse reactions [3]. IV iron allows for administration of larger doses with better tolerability and is considered to be superior to oral iron in patients with CKD [41].
Although rare, IV iron administration may be associated with an increased risk of iron overload, which could potentially lead to organ dysfunction in patients with or without ESRD, although end-organ damage due to IV iron has not been demonstrated in clinical studies [42]. Iron overload can also increase infection risk and worsen CKD-associated inflammation, while inflammation can exacerbate oxidative stress caused by IV iron [42, 43]. Previous reports of hypersensitivity with IV iron were largely during the use of high-molecular-weight iron dextrans that are no longer commercially available [44, 45]. IV iron is burdensome in patients with NDD-CKD because of the need for IV access and a transfusion clinic [46].

Erythropoiesis-Stimulating Agents

ESAs trigger EPO production to increase Hb and improve anemia [3]. Although ESAs reduce the adverse impact of anemia on morbidity and HR-QOL [47], safety concerns regarding the potential increased risk of cardiovascular events with increased ESA doses (due to poor response or a higher Hb target) have led to reductions in the prescribed ESA dose, increased use of RBC transfusion/IV iron, and uncertainty regarding optimal target Hb [4]. Consequently, regulatory authorities increasingly require detailed safety data for ESAs. Other considerations for ESA use include parenteral administration, cold storage, expense, and the generation of neutralizing anti-EPO antibodies, which may cause pure red cell aplasia [4].

Impact of ESA-Mediated Anemia Correction

Hemoglobin normalization in patients with CKD is currently not recommended because of safety concerns related to ESA dosage [48]. Some studies show cardiovascular benefits in treating to a lower Hb target while others describe poor cardiovascular outcomes with a physiologically normal or supraphysiologic Hb target, rendering the optimal target Hb uncertain [3, 4, 30]. Higher ESA dose (rather than higher Hb) may cause adverse effects, as ESRD patients who maintain high Hb (> 12 g/dl) without ESA therapy do not show increased mortality compared with other patients on dialysis [49]. Current guidelines recommend a target Hb ≤ 11.5 g/dl [3].
Anemia correction with ESAs may provide improvement in cardiovascular parameters, including ejection fraction, left ventricular (LV) mass index, and LV wall thickness [22, 50]. In patients with NDD-CKD, the risk of renal events (i.e., progression to renal replacement therapy, doubling of serum creatinine, or decline in eGFR to < 6 ml/min/1.73m2) was significantly lower in those with Hb target of ≥ 11 g/dl versus < 11 g/dl [51]. However, the ACORD, CHOIR, and CREATE studies in patients with NDD-CKD showed no advantage with a high (13.0–15.0 g/dl) versus low (10.5–11.5 g/dl) Hb target in the risk for LVH [52] or cardiovascular events (including sudden death, stroke, transient ischemic attack, MI, acute heart failure, hospitalization for angina pectoris, cardiac arrhythmia, or congestive heart failure, or complication of peripheral vascular disease) [20, 53]. Additionally, in a subanalysis of the TREAT trial, poor initial response to ESA therapy (and consequently higher doses of ESA) in patients with NDD-CKD and type 2 diabetes was associated with increased risks of all-cause death (HR 1.41, 95% CI 1.12–1.78) and adverse cardiovascular events (HR 1.31, 95% CI 1.09–1.59) compared with patients with better response to ESA [54]. Due to greater risks for death, MACE, and stroke with target Hb ≥ 13 g/dl [20, 21], the US Food and Drug Administration (FDA) recommends that ESA dosing be individualized to the lowest dose necessary to reduce RBC transfusion requirements rather than to a specific target Hb [48]. Notably, following the FDA communication, there was a 59%–74% decrease in the prescribing of ESAs despite stable anemia prevalence rates [55]. However, there was no corresponding reduction in the rate of mortality or MACE [56].

Impact of ESAs on HR-QOL

Although benefits are reported often, significant improvements in HR-QOL following ESA treatment of anemia in patients with CKD are inconsistent. ESA therapy was associated with significant improvements in fatigue, vitality, mental health/emotional well-being, and overall physical health in patients with NDD-CKD [20]. Correction of anemia to a target Hb of 13–15 g/dl improved HR-QOL in patients with CKD with or without diabetes [52, 53] with improvements in several subscales of the Short Form 36 health survey versus a target Hb of 10.5–11.5 g/dl [53]. In contrast, a meta-analysis showed that ESA therapy to obtain higher Hb targets (10.2–13.6 g/dl) does not improve HR-QOL [57]. In patients with CKD on dialysis, ESA therapy is associated with better overall HR-QOL and lower costs and healthcare resource utilization compared with no ESA therapy, although there appears to be minimal benefit with higher Hb targets [58]. Partial correction of anemia with ESAs in dialysis patients has been shown to reduce fatigue and improve exercise tolerance and general well-being, while high-dose ESA was associated with increased cardiovascular risk that negatively impacted HR-QOL, thereby resulting in only a modest overall improvement [59, 60].

Red Blood Cell Transfusion

Before ESA availability, frequent RBC transfusion was the primary means of correcting CKD anemia [47]. Currently, ~20% of patients with NDD-CKD receive RBC transfusions [61]; however, blood volume overload, hyperkalemia, iron overload, blood-borne infections, fever, or allosensitization may occur [3]. Given the burdens associated with RBC transfusion, clinicians should consider alternative treatments for anemia in CKD [61]. However, RBC transfusion may be the only available option in some patients in whom ESAs are not recommended, for example, cancer patients with non-chemotherapy-associated anemia (except for selected patients with myelodysplastic syndrome) [62].

Special Populations

Elderly Patients

The prevalence of cardiovascular conditions increases in elderly patients with anemia of CKD [1]. Indeed, CKD, anemia, and mobility limitation are important prognostic indicators of mortality risk in elderly patients [63]. Older patients with CKD have higher rates of inflammatory conditions, nutritional deficiencies, and cardiovascular comorbidities, as well as increased hepcidin levels [64], potentially complicating iron and/or ESA therapy. In addition, Hb decreases with age because of reduced erythropoiesis, so the optimal target Hb in elderly patients may be lower [64].

Diabetes

Type 2 diabetes frequently contributes to CKD development and may also increase the risk of anemia in CKD [65]. Diabetes is an inflammatory condition exacerbated by hyperglycemia and other inflammatory disorders, including obesity, arterial hypertension, and dyslipidemia; this increased inflammation is thought to cause EPO deficiency in patients with diabetes [6668]. Deficiencies in EPO and iron, as well as hyporesponsiveness to EPO, are the main mechanisms for anemia development in patients with diabetic kidney disease [69]. In patients with diabetes, anemia is generally more severe, occurs at an earlier stage of CKD, and is associated with a potentially greater risk of cardiovascular disease [70]. Additionally, diabetic macrovascular complications also contribute to the development of atherosclerosis [71], which can further complicate anemia management. However, despite the increased risk of adverse clinical outcomes in patients with diabetes and anemia, there is often clinical inertia regarding initiating IV iron or ESA therapy in these patients [72]. In patients with comorbid diabetes, treatment with the ESA darbepoetin alfa showed no reduction in the risk of composite outcomes (death or cardiovascular event and death or renal event) and an increased risk of stroke versus placebo [21]. In this study, patients with poor initial response to ESA therapy (who received higher ESA doses to meet Hb targets) had increased risks of all-cause mortality (HR 1.41, 95% CI 1.12–1.78) and cardiovascular events (HR 1.31, 95% CI 1.09–1.59) than those with better initial response [54]. This indicates that some patients with diabetes and anemia may benefit from alternative therapies, eliminating the need for ESA dose escalation in those with poor initial response to ESA therapy.

End-Stage Renal Disease

In patients with stage 3 CKD, those who develop anemia have more rapid progression to stage 4 and 5 CKD [73]. Dialysis plays a key role in ESRD management, but HR-QOL for patients with DD-CKD remains a concern, suggesting the need for a more patient-centric assessment [74]. In addition to blood loss associated with hemodialysis, complications of severe anemia contribute significantly to a decreased HR-QOL and increased dependence on RBC transfusion [75]. Iron overload is another concern and was observed in 84% of patients with DD-CKD treated with ESAs and IV iron [76]. Similar to patients with NDD-CKD, adverse outcomes occur in patients with DD-CKD, with higher mortality rates and no difference in cardiovascular events when epoetin was used to target higher versus lower hematocrit [77]. Notably, attenuation of CKD progression has not been shown with ESA therapy.

Kidney Transplantation

Anemia prevalence decreases following kidney transplant, from 71% pre-transplant to 51% at 6 months and 37% at 2 years post-transplant. However, post-transplant anemia does occur [78]. In kidney transplant recipients, lower Hb is a predictor for a return to dialysis, graft failure, subsequent kidney transplant, reduced LV mass index, or death [78, 79]. ESA use to target high Hb (12.5–13.5 g/dl) appears to attenuate the decline of kidney function compared with low Hb (10.5–11.5 g/dl) after 3 years of follow-up in kidney transplant recipients [80]. Of note, patients with ESA hyporesponsiveness before kidney transplant remained hyporesponsive following transplant [81], indicating a need for new therapies to treat anemia in this subpopulation.

Emerging Alternatives

Given the inherent limitations of the current standard of care, new effective and tolerable treatment options for CKD anemia are needed. One particularly promising class of agents in development is hypoxia-inducible factor-prolyl hydroxylase (HIF-PH) inhibitors.

HIF-PH Inhibitors

Hypoxia-inducible factor (HIF) regulates gene expression in response to hypoxia, including genes involved in erythropoiesis and iron metabolism, promoting iron absorption, iron transport, and heme synthesis (Fig. 1) [37]. Notably, work on the discovery of HIF and its mechanism of action received the 2019 Nobel Prize in Physiology or Medicine. Under normoxic conditions, HIF-PH enzymes promote HIF degradation; thus, selective HIF stabilization with HIF-PH inhibitors is an innovative approach for treating anemia of CKD [36, 82]. Several HIF-PH inhibitors are currently under development (Table 2). HIF-PH inhibitors are orally administered, and significantly lower EPO levels are induced compared with the supraphysiologic levels typically attained with ESA therapy (Fig. 2) [82]. Animal studies have shown that HIF-PH inhibitors stimulate EPO expression in the kidneys and liver, increasing Hb levels in models of anemia of CKD, including 5/6th nephrectomized rats [83, 84]. HIF-PH inhibitors have also been shown to decrease hepcidin, which may allow patients to mobilize iron stores and lessen iron supplementation needs. Additionally, HIF stabilization should increase gastrointestinal iron absorption through increased expression of divalent metal transporter-1 and duodenal cytochrome B [85].
Table 2
Phase 2 and 3 clinical trials of hypoxia-inducible factor prolyl-hydroxylase inhibitors
Trial identifier
Participants
Nb
Study design
Comparator
Location
Treatment duration
Roxadustata
 Patients with DD-CKD
  NCT01596855 [86]
ESRD, hemodialysis, Hb 9–12 g/dl, stable epoetin 7 weeks
87
Phase 2 RCT OL
Epoetin alfa
China
6 weeks
  NCT01147666 [87]
ESRD, maintenance hemodialysis ≥ 4 months, Hb 9.0–13.5 g/dl for 8 weeks, epoetin alfa and intravenous iron 4 weeks
144
Phase 2 RCT OL
Epoetin alfa
US
6 weeks
  NCT01414075 [88]
Incident dialysis (2 weeks–4 months), Hb ≤ 10 g/dl, ferritin 50–300 ng/ml, TSAT 10%–30%, ESA-naïve, no intravenous iron ≥ 4 weeks
60
Phase 2b RCT OL
None
US, Russia, Hong Kong
12 weeks
  NCT02652806 [35]
ESRD, dialysis ≥ 16 week, Hb 9.0–12.0 g/dl, stable epoetin alfa ≥ 6 weeks
305
Phase 3 RCT OL
Epoetin alfa
China
26 weeks
  NCT02779764 [89]
Hemodialysis 3 times/weeks for ≥ 12 weeks, ESA ≥ 8 weeks, mean of 2 latest Hb levels 10–12 g/dl, TSAT ≥ 20% or ferritin ≥ 100 ng/ml
164
Phase 3
None
Japan
52 weeks
  NCT02780141 [89]
Hemodialysis ≥ 1 time/weeks, ESA-naïve, mean of 2 latest Hb levels ≤ 10 g/dl, TSAT ≥ 5% or ferritin ≥ 30 ng/ml
75
Phase 3 RCT OL
None
Japan
24 weeks
  NCT02273726 (SIERRAS) [90]
ESRD, dialysis ≥ 3 months, Hb 8.5–12.0 g/dl, ferritin ≥ 100 ng/ml, TSAT ≥ 20%, ESA ≥ 4 weeks
741
Phase 3 RCT OL
Epoetin alfa
US
52 weeks to 3 years
  NCT02174731 (ROCKIES) [91]
Hemodialysis or peritoneal dialysis; Hb < 12.0 g/dl in those on ESA, < 10 g/dl in those not on ESA; ferritin ≥ 100 ng/ml; TSAT ≥ 20%
2133
Phase 3 RCT OL
Epoetin alfa
North America, Asia, Australia, EU, India, South America
52 weeks to 4 years
  NCT02278341 (PYRENEES) [92]
Stable hemodialysis or peritoneal dialysis, Hb 9.5–12 g/dl, epoetin alfa or darbepoetin alfa ≥ 8 weeks
836
Phase 3 RCT OL
ESA (epoetin alfa or darbepoetin alfa)
EU
52–104 weeks
  NCT02052310 (HIMALAYAS) [93]
ESRD, incident dialysis (2 weeks–4 months)
1043
Phase 3 RCT OL
Epoetin alfa
US, Asia, EU, South America
52 weeks to 3 years
 Patients with NDD-CKD
  NCT01599507 [86]
NDD-CKD (eGFR < 60 ml/min/1.73 m2), Hb < 10 g/dl
91
Phase 2 RCT DB
 
China
8 weeks
  NCT01244763 [94]
NDD-CKD (eGFR 15–59 ml/min/1.73 m2), Hb < 10.5 g/dl, ferritin > 30 ng/ml, TSAT ≥ 5%, no ESA use ≤ 12 weeks
145
Phase 2 RCT OL
None
US
16 or 24 weeks
  NCT00761657 [95]
NDD-CKD stage 3–4 (eGFR 15–59 ml/min/1.73 m2), Hb < 11 g/dl
117
Phase 2a RCT
 
US
4 weeks
  NCT02652819 [96]
NDD-CKD stage 3–5, Hb 7– < 10 g/dl, no ESA use ≤ 5 weeks
154
Phase 3 RCT DB followed by OL extension
 
China
8 weeks (RCT); 18 weeks (OL)
  NCT01750190 (ANDES) [97]
NDD-CKD stage 3–5
922
Phase 3 RCT DB
 
US, Asia, Australia, South America
 < 52 weeks to 3 years
  NCT01887600 (ALPS) [92]
NDD-CKD stage 3–5 (eGFR < 60 ml/min/1.73 m2), Hb ≤ 10 g/dl, ferritin ≥ 30 ng/ml, TSAT ≥ 5%, ESA-naïve
594
Phase 3 RCT DB
 
EU
52–104 weeks
  NCT02174627 (OLYMPUS) [98]
NDD-CKD stage 3–5 (eGFR < 60 ml/min/1.73 m2), Hb ≤ 10 g/dl, ferritin ≥ 50 ng/ml, TSAT ≥ 15%, ESA-naïve
2781
Phase 3 RCT DB
 
North America, Asia, EU, India, South America
52 weeks
Daprodustata
 Patients with DD-CKD
  NCT02019719 [99]
Hemodialysis ≥ 8 weeks, Hb 9.5–12.0 g/dl, ferritin ≥ 100 μg/l, TSAT ≥ 20%, stable ESA use ≥ 4 weeks
97
Phase 2, RCT DB
 
Japan
4 weeks
  NCT01587924 [100]
Hemodialysis ≥ 8 weeks, Hb 9.5–12.0 g/dl, ferritin ≥ 40 ng/ml, stable ESA use ≥ 4 weeks
83
Phase 2a RCT DB
ESA
US, Canada, EU
4 weeks
  NCT02075463 [101]
Stable hemodialysis ≥ 12 weeks, ESA hyporesponsiveness, ferritin ≥ 100 ng/ml, TSAT ≥ 20%
15
Phase 2a OL
None
US
16 weeks
  NCT01977482 [102]
Adequate hemodialysis, Hb 9–11.5 g/dl, ferritin < 100 ng/ml, TSAT < 12%– > 57%, stable ESA use ≥ 4 weeks
177
Phase 2b RCT DB
ESA
US, Australia, EU, Canada, Asia
24 weeks
  NCT02829320 [103]
Hemodialysis (newly initiated < 12 weeks and ESA-naïve or maintenance ≥ 12 weeks and no ESA use ≥ 8 weeks), Hb ≥ 8– < 10 g/dl, ferritin ≥ 100 ng/ml
28
Phase 3 RCT OL
None
Japan
24 weeks
 Patients with NDD-CKD
  NCT01977573 [104]
NDD-CKD stage 3–5; Hb 8–11 g/dl (ESA-naïve), 9–11.5 g/dl (ESA users); for ESA users, stable ESA use ≥ 4 weeks
252
Phase 2 RCT
ESA
US, Australia, EU, Canada, Asia
24 weeks
  NCT01587898 [100]
NDD-CKD stage 3–5, Hb 8.5–11.0 g/dl, ferritin ≥ 40 ng/ml or TSAT in reference range, no ESA use ≥ 7 weeks
73
Phase 2a RCT DB
 
US, Canada, EU
4 weeks
 Patients with NDD-CKD or DD-CKD
  NCT01047397 [105]
CKD stage 3–4 (eGFR 15–59 ml/min/1.73 m2), CKD stage 5 (eGFR 10– < 15 ml/min/1.73 m2), or CKD stage 5d (eGFR 10– < 15 ml/min/1.73 m2 and hemodialysis); ESA-naïve with Hb ≤ 11 g/dl or no ESA use ≥ 7 days
107
Phase 2a RCT
 
Australia, India, Russia
28 days
Vadadustata
 Patients with DD-CKD
 NCT02260193 [106]
Maintenance hemodialysis thrice weekly ≥ 3 months, epoetin alfa and intravenous iron ≥ 3 months
94
Phase 2 OL
None
US
16 weeks
 Patients with NDD-CKD
  NCT01906489 [107]
NDD-CKD stage 3a–5, ferritin level ≥ 50 ng/ml with TSAT ≥ 18% or a ferritin level ≥ 100 ng/ml regardless of TSAT
210
Phase 2 RCT DB
 
US
20 weeks
  NCT01381094 [108]
CKD stage 3 or 4 (eGFR 30–59 or 15–29 ml/min/1.73 m2), no ESA ≥ 11 weeks, Hb ≤ 10.5 g/dl, ferritin ≥ 50 ng/ml, TSAT ≥ 20%
93
Phase 2a, RCT
 
US
6 weeks
Molidustata
 Patients with DD-CKD
  NCT01975818 (DIALOGUE 4) [109]
DD-CKD, Hb 9.0–11.5 g/dl, stable epoetin use ≥ 8 weeks
199
Phase 2b RCT OL
Epoetin alfa/beta
US, Japan
16 weeks
  NCT02064426 (DIALOGUE 5) [110]
DD-CKD (from DIALOGUE 4)
88
OL extension of DIALOGUE 4
Epoetin alfa/beta
US, Japan
 ≤ 36 months
 Patients with NDD-CKD
  NCT02021370 (DIALOGUE 1) [109]
NDD-CKD (ESA-naïve eGFR < 60 ml/min/1.73 m2), Hb < 10.5 g/dl, ESA-naïve or no ESA use ≥ 8 weeks
121
Phase 2b RCT DB
 
EU, Asia–Pacific
16 weeks
  NCT02021409 (DIALOGUE 2) [109]
NDD-CKD (eGFR < 60 ml/min/1.73 m2), Hb 9–12 g/dl, stable darbepoetin use ≥ 8 weeks
124
Phase 2b RCT OL
Darbepoetin
EU, Asia–Pacific
16 weeks
  NCT02055482 (DIALOGUE 3) [110]
NDD-CKD (from DIALOGUE 1 and 2)
164
OL extension of DIALOGUE 1 and 2
Darbepoetin
EU, Asia–Pacific
 ≤ 36 months
Enarodustata
 Patients with DD-CKD
  JapicCTI-152892 [111]
Hemodialysis or hemodiafiltration 3 times per weeks ≥ 12 weeks, ESA therapy ≥ 4 weeks, mean Hb at screening and 2 weeks later 9.5–12.0 g/dl with absolute difference of ≤ 1.0 g/dl, TSAT > 20% or ferritin > 75 ng/ml
85
Phase 2b RCT DB followed by OL extension
 
Japan
6 weeks (RCT); 24 weeks (OL)
 Patients with NDD-CKD
  JapicCTI-152881 [112]
CKD not on dialysis (eGFR < 60 ml/min/1.73 m2), mean Hb 8.0–10.5 g/dl for correction group (ESA-naïve: no ESA ≥ 12 weeks) and 9.5–12.0 g/dl for conversion group (ESA-treated: stable ESA ≥ 8 weeks)
201
Phase 2b RCT DB followed by OL extension
 
Japan
6 weeks (RCT); 24 weeks (OL)
Desidustata
 Patients with NDD-CKD
  CTRI/2017/05/008534 [113]
NDD-CKD stage 1–4, Hb 6.5–11 g/dl, ferritin 100–1000 μg/l or TSAT ≥ 20%, body weight ≥ 45 kg
117
Phase 2 RCT DB
 
India
6 weeks
CKD chronic kidney disease, DB double blind, DD dialysis-dependent, eGFR estimated glomerular filtration rate, ESA erythropoiesis-stimulating agent, ESRD end-stage renal disease, EU European Union, Hb hemoglobin, NDD non-dialysis-dependent, OL open-label, RCT randomized controlled trial, TSAT transferrin saturation
aHalf-life for roxadustat: 11.4–14.7 h [114116]; daprodustat: 0.9–2.3 h [117]; vadadustat: 4.7–9.1 h [118]; molidustat: mean, 4.6–10.4 h [119]; enarodustat: not available; desidustat: mean, 6.9–11.4 h [120]
bNumber randomized

Approved HIF-PH Inhibitors

Roxadustat (FG-4592) was the first-in-class HIF-PH inhibitor approved in Japan for the treatment of anemia in patients with DD-CKD [121] and in China for patients with DD-CKD or NDD-CKD [122]. Daprodustat (GSK1278863) and vadadustat (AKB-6548) are also now approved in Japan for the treatment of anemia in patients with DD-CKD or NDD-CKD [123, 124]. All three HIF-PH inhibitors effectively stimulate EPO production in patients with anemia of CKD, providing dose-dependent increases in Hb and reductions in hepcidin levels, and thus improving total iron binding capacity (TIBC) [35, 9093, 9698, 103, 125127].
In NDD-CKD patients, roxadustat was associated with superior and/or statistically significant Hb response rates and changes from baseline compared with placebo in a Chinese phase 3 randomized study [96] and in preliminary results from three international phase 3 studies [92, 97, 98]. In these studies, roxadustat was also associated with a reduced risk of rescue therapy (ESA or IV iron) and RBC transfusion [98] and reduced hepcidin levels compared with placebo (between group difference −50 ng/ml) [96]. Interim data from a phase 3 study showed that roxadustat was noninferior to darbepoetin alfa regarding Hb response in NDD-CKD patients [125]. Preliminary data from a Japanese phase 3 study showed that vadadustat was as effective as darbepoetin alfa in maintaining Hb levels in both ESA-naïve and ESA-converted NDD-CKD patients with anemia [126].
In both ESA-naïve and -experienced DD-CKD patients with anemia, roxadustat demonstrated non-inferiority or superiority in increasing Hb from baseline versus epoetin alfa or darbepoetin alfa in a Chinese phase 3 study [35] and in preliminary data from four international phase three studies [9093]. Greater decreases in hepcidin from baseline were also observed with roxadustat versus epoetin alfa [35]. In a phase 3 Japanese study in ESA-naïve hemodialysis patients, daprodustat effectively corrected and maintained Hb levels within the target range (10–12 g/dl), decreased hepcidin levels, and increased TIBC [103]. Similarly, preliminary data demonstrated that vadadustat was as effective as darbepoetin alfa in maintaining Hb levels within the target range in Japanese patients on maintenance hemodialysis and resulted in reduced hepcidin levels and increased TIBC over 24 weeks, which was not observed in the darbepoetin alfa group [127].
HIF-PH inhibitors were well tolerated in phase 3 clinical studies, and adverse events (AEs) were consistent with those expected in a CKD population [35, 92, 96, 126, 127]. The most common AEs with roxadustat were hyperkalemia and metabolic acidosis in NDD-CKD patients [96] and hyperkalemia in DD-CKD patients [35]. Additionally, preliminary data from two further international phase 3 studies reported the most common AEs with roxadustat to be ESRD, urinary tract infection, pneumonia, and hypertension in NDD-CKD patients [128] and diarrhea in DD-CKD patients [129]. The most commonly reported AE with daprodustat in DD-CKD patients was nasopharyngitis [103]. For vadadustat, these were nasopharyngitis, diarrhea, and constipation in NDD-CKD patients [126] and nasopharyngitis, constipation, and shunt stenosis in DD-CKD patients [127].
Preliminary results from a pooled safety analysis of NDD-CKD or stable DD-CKD patients with anemia indicated a similar or reduced risk of MACE and MACE plus heart failure or unstable angina requiring hospitalization (MACE+)with roxadustat versus placebo and epoetin alfa, respectively [130]. In incident DD-CKD patients with anemia, the HRs for MACE and MACE+ were 0.70 (95% CI 0.51–0.97, P = 0.03) and 0.66 (95% CI 0.5–0.89, P = 0.005), respectively, with roxadustat versus epoetin alfa [130]. Further analyses are needed to confirm these initial safety findings.

HIF-PH Inhibitors in Development

Several other HIF-PH inhibitors are in development, with data available for molidustat (BAY 85-3934), enarodustat (JTZ-951), and desidustat (Zyan1) (Table 2). These studies show dose-dependent Hb increases and maintenance of Hb (in NDD-CKD) and maintenance of Hb (in DD-CKD) for molidustat [109], enarodustat [111, 112], and desidustat [113]. However, high Hb or a rapid rate of increase led to high incidences of early discontinuation from some studies of molidustat [109]. In the long-term extension studies DIALOGUE 3 and DIALOGUE 5, Hb was maintained in the target range (10–12 g/dl) for up to 36 months with molidustat, with a similar effect to darbepoetin or epoetin [110]. Increased TIBC and/or decreased hepcidin and/or ferritin was observed with these agents, which were generally well tolerated [109, 112, 113]. Furthermore, animal studies have indicated that prolonged exposure to roxadustat is not associated with pro-oncogenic activity [131, 132]. However, long-term clinical data are needed to confirm the safety of HIF-PH inhibitors regarding to cardiovascular events and carcinogenesis.

Potential for Clinical Use of HIF-PH Inhibitors

HIF-PH inhibitors may present several practical advantages for patients with anemia of CKD. In addition to their oral route of administration, HIF-PH inhibitors may provide closer to physiologic EPO levels than the intermittent high levels attained with ESA therapy [87, 95]. Beyond erythropoiesis stimulation, HIF-PH inhibitors may improve iron homeostasis [133] and therefore reduce patients’ iron supplementation needs, thus potentially reducing costs and medication burden. Although data on the cost effectiveness of HIF-PH inhibitors are limited, a meta-analysis conducted to evaluate the cost effectiveness of roxadustat in Chinese patients with NDD-CKD confirmed that roxadustat was cost effective compared with placebo [134].
Evidence suggests that HIF-PH inhibitors may be efficacious without increasing inflammatory status [88], which could benefit patients with inflammation, associated with diabetic and non-diabetic kidney disease as well as those with acute inflammation (e.g., associated with infection). Although clinical data in patients who are ESA hyporesponsive are limited, key studies included patients with moderate inflammation, which is associated with reduced responsiveness to ESA therapy [135]. In the Chinese phase 3 study of roxadustat in patients with DD-CKD, similar increases in Hb levels were observed in patients with normal and elevated C-reactive protein levels (≤ 4 and > 4 mg/l) [35]. In addition, preliminary phase 3 data showed greater mean changes in Hb in patients with elevated high-sensitivity C-reactive protein levels receiving roxadustat versus epoetin alfa (DD-CKD) [91] or placebo (NDD-CKD) [98]. In these patients with moderate inflammation, who are potentially hyporesponsive to ESA therapy, HIF-PH inhibitors may be an effective alternative that avoids the need for high-dose ESA therapy. Further studies are needed to confirm the efficacy of HIF-PH inhibitors in patients who are ESA hyporesponsive. Finally, HIF-PH inhibitors may confer a reduced risk of cardiovascular events compared with ESAs in incident dialysis patients as a preliminary phase 3 pooled analysis showed a lower risk of MACE and MACE+ with roxadustat versus epoetin alfa [130]. Further studies are needed to confirm the practical benefits of HIF-PH inhibitors in patients with anemia of CKD.
Because HIF transcription factors regulate many biologic processes, there was concern that HIF-PH inhibitors may adversely affect cholesterol metabolism [136]. Based on animal studies, constitutive HIF-2 activation may theoretically suppress hepatic fatty acid β-oxidation and lipid synthesis and increase lipid storage capacity [136]. However, clinical studies showed reductions in total and low-density lipoprotein cholesterol (LDL-C) with roxadustat over 19–24 weeks [87, 94] and daprodustat over 24 weeks [103] as well as no changes in serum lipids with vadadustat over 16 or 20 weeks [106, 107] and only small changes in LDL-C with molidustat over 16 weeks [109]. Roxadustat phase 3 data showed decreases in low-density lipoprotein cholesterol versus placebo (NDD-CKD patients) [96] or versus ESA (DD-CKD patients) [35]. One potential mechanism for this reduction in serum cholesterol with roxadustat is thought to be a HIF-dependent decrease in 3-hydroxy-3-methylglutaryl coenzyme A reductase levels, a rate-limiting enzyme in the cholesterol biosynthesis pathway [137].

At-Home Anemia Management

At-home care of CKD is one of the goals outlined in the recent Executive Order, Advancing American Kidney Health, which aims to improve the diagnosis and treatment of CKD [138]. Compared with conventional hemodialysis, at-home hemodialysis benefits include reductions in LV mass and hypertension and increased HR-QOL, although there are no observed differences in anemia management [139, 140].
Because they are orally administered, HIF-PH inhibitors may confer advantages for at-home CKD care. In ESRD patients receiving peritoneal dialysis, the more common modality for at-home dialysis, roxadustat increased Hb to within the target range [141], and daprodustat pharmacokinetics were similar in patients receiving peritoneal dialysis or in-center hemodialysis, while Hb was maintained in those receiving peritoneal dialysis [142].

Conclusions

Anemia of CKD represents a considerable burden to both patients and the healthcare system. Although effective, the current standard of care is associated with inherent practical difficulties and safety concerns, including the increased risk of cardiovascular events and mortality. HIF-PH inhibitors may offer advantages over ESAs through more physiologic and effective means of treating anemia of CKD.

Acknowledgements

Funding

This review, the Rapid Service, and Open Access Fees were funded by AstraZeneca.

Editorial Assistance

Sarah Greig, PhD (Auckland, NZ), and Meri D. Pozo, PhD, CMPP (New York, NY, USA), of inScience Communications, Springer Healthcare provided editorial support, which was funded by AstraZeneca.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Authorship Contributions

All authors wrote the first draft of the manuscript and participated in subsequent drafts, approved the submission of the manuscript, and are fully accountable for all aspects of the work.

Disclosures

Ramy M. Hanna and Elani Streja have nothing to disclose. Kamyar Kalantar-Zadeh reports personal fees from Abbott, AbbVie, Alexion, AMAG Pharma, Amgen, AstraZeneca, AVEO, Baxter, Chugai, Fresenius Medical Services, Genentech, Haymarket, Hospira, Fresenius Kabi USA, Keryx, Novartis, PCORI, Pfizer, Relypsa, Resverlogix, Sandoz, Sanofi, Shire, Vifor, UpToDate, and grants and personal fees from National Institutes of Health. None of the authors received honoraria for this work.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial 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-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat St Peter WL, Guo H, Kabadi S, et al. Prevalence, treatment patterns, and healthcare resource utilization in Medicare and commercially insured non-dialysis-dependent chronic kidney disease patients with and without anemia in the United States. BMC Nephrol. 2018;19:67.PubMedCrossRefPubMedCentral St Peter WL, Guo H, Kabadi S, et al. Prevalence, treatment patterns, and healthcare resource utilization in Medicare and commercially insured non-dialysis-dependent chronic kidney disease patients with and without anemia in the United States. BMC Nephrol. 2018;19:67.PubMedCrossRefPubMedCentral
2.
Zurück zum Zitat Mikhail A, Brown C, Williams JA, et al. Renal association clinical practice guideline on Anaemia of Chronic Kidney Disease. BMC Nephrol. 2017;18:345.PubMedCrossRefPubMedCentral Mikhail A, Brown C, Williams JA, et al. Renal association clinical practice guideline on Anaemia of Chronic Kidney Disease. BMC Nephrol. 2017;18:345.PubMedCrossRefPubMedCentral
3.
Zurück zum Zitat Kidney Disease Improving Global Outcomes. KDIGO clinical practice guideline for anemia in chronic kidney disease. Kidney Int Suppl. 2012;2:279–335.CrossRef Kidney Disease Improving Global Outcomes. KDIGO clinical practice guideline for anemia in chronic kidney disease. Kidney Int Suppl. 2012;2:279–335.CrossRef
4.
Zurück zum Zitat Bonomini M, Del Vecchio L, Sirolli V, Locatelli F. New treatment approaches for the anemia of CKD. Am J Kidney Dis. 2016;67:133–42.PubMedCrossRef Bonomini M, Del Vecchio L, Sirolli V, Locatelli F. New treatment approaches for the anemia of CKD. Am J Kidney Dis. 2016;67:133–42.PubMedCrossRef
6.
Zurück zum Zitat Hill NR, Fatoba ST, Oke JL, et al. Global prevalence of chronic kidney disease: a systematic review and meta-analysis. PLoS ONE. 2016;11:e0158765.PubMedCrossRefPubMedCentral Hill NR, Fatoba ST, Oke JL, et al. Global prevalence of chronic kidney disease: a systematic review and meta-analysis. PLoS ONE. 2016;11:e0158765.PubMedCrossRefPubMedCentral
9.
Zurück zum Zitat London GM. Left ventricular alterations and end-stage renal disease. Nephrol Dial Transpl. 2002;17(Suppl 1):29–36.CrossRef London GM. Left ventricular alterations and end-stage renal disease. Nephrol Dial Transpl. 2002;17(Suppl 1):29–36.CrossRef
10.
Zurück zum Zitat Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation. 2003;108:2154–69.PubMedCrossRef Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation. 2003;108:2154–69.PubMedCrossRef
11.
12.
Zurück zum Zitat Toft G, Heide-Jorgensen U, van Haalen H, et al. Anemia and clinical outcomes in patients with non-dialysis dependent or dialysis dependent severe chronic kidney disease: a Danish population-based study. J Nephrol. 2020;33:147–56.PubMedCrossRef Toft G, Heide-Jorgensen U, van Haalen H, et al. Anemia and clinical outcomes in patients with non-dialysis dependent or dialysis dependent severe chronic kidney disease: a Danish population-based study. J Nephrol. 2020;33:147–56.PubMedCrossRef
13.
Zurück zum Zitat Iimori S, Naito S, Noda Y, et al. Anaemia management and mortality risk in newly visiting patients with chronic kidney disease in Japan: The CKD-ROUTE study. Nephrology. 2015;20:601–8.PubMedCrossRef Iimori S, Naito S, Noda Y, et al. Anaemia management and mortality risk in newly visiting patients with chronic kidney disease in Japan: The CKD-ROUTE study. Nephrology. 2015;20:601–8.PubMedCrossRef
14.
Zurück zum Zitat Abramson JL, Jurkovitz CT, Vaccarino V, Weintraub WS, McClellan W. Chronic kidney disease, anemia, and incident stroke in a middle-aged, community-based population: the ARIC Study. Kidney Int. 2003;64:610–5.PubMedCrossRef Abramson JL, Jurkovitz CT, Vaccarino V, Weintraub WS, McClellan W. Chronic kidney disease, anemia, and incident stroke in a middle-aged, community-based population: the ARIC Study. Kidney Int. 2003;64:610–5.PubMedCrossRef
15.
Zurück zum Zitat Vlagopoulos PT, Tighiouart H, Weiner DE, et al. Anemia as a risk factor for cardiovascular disease and all-cause mortality in diabetes: the impact of chronic kidney disease. J Am Soc Nephrol. 2005;16:3403–10.PubMedCrossRef Vlagopoulos PT, Tighiouart H, Weiner DE, et al. Anemia as a risk factor for cardiovascular disease and all-cause mortality in diabetes: the impact of chronic kidney disease. J Am Soc Nephrol. 2005;16:3403–10.PubMedCrossRef
16.
Zurück zum Zitat Lee G, Choi S, Kim K, et al. Association of hemoglobin concentration and its change with cardiovascular and all-cause mortality. J Am Heart Assoc. 2018;7:e007723.PubMedPubMedCentral Lee G, Choi S, Kim K, et al. Association of hemoglobin concentration and its change with cardiovascular and all-cause mortality. J Am Heart Assoc. 2018;7:e007723.PubMedPubMedCentral
17.
Zurück zum Zitat Kim-Mitsuyama S, Soejima H, Yasuda O, et al. Anemia is an independent risk factor for cardiovascular and renal events in hypertensive outpatients with well-controlled blood pressure: a subgroup analysis of the ATTEMPT-CVD randomized trial. Hypertens Res. 2019;42:883–91.PubMedCrossRef Kim-Mitsuyama S, Soejima H, Yasuda O, et al. Anemia is an independent risk factor for cardiovascular and renal events in hypertensive outpatients with well-controlled blood pressure: a subgroup analysis of the ATTEMPT-CVD randomized trial. Hypertens Res. 2019;42:883–91.PubMedCrossRef
18.
Zurück zum Zitat Zoppini G, Targher G, Chonchol M, et al. Anaemia, independent of chronic kidney disease, predicts all-cause and cardiovascular mortality in type 2 diabetic patients. Atherosclerosis. 2010;210:575–80.PubMedCrossRef Zoppini G, Targher G, Chonchol M, et al. Anaemia, independent of chronic kidney disease, predicts all-cause and cardiovascular mortality in type 2 diabetic patients. Atherosclerosis. 2010;210:575–80.PubMedCrossRef
19.
Zurück zum Zitat Fishbane S, Spinowitz B. Update on anemia in ESRD and earlier stages of CKD: core curriculum 2018. Am J Kidney Dis. 2018;71:423–35.PubMedCrossRef Fishbane S, Spinowitz B. Update on anemia in ESRD and earlier stages of CKD: core curriculum 2018. Am J Kidney Dis. 2018;71:423–35.PubMedCrossRef
20.
Zurück zum Zitat Singh AK, Szczech L, Tang KL, et al. Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med. 2006;355:2085–98.PubMedCrossRef Singh AK, Szczech L, Tang KL, et al. Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med. 2006;355:2085–98.PubMedCrossRef
21.
Zurück zum Zitat Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med. 2009;361:2019–32.PubMedCrossRef Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med. 2009;361:2019–32.PubMedCrossRef
22.
Zurück zum Zitat Mukhopadhyay P, Sinha U, Banerjee J, Choudhury AR, Philips CA. The effects in correction of anaemia in chronic kidney disease with erythropoietin therapy–preference to cardiovascular, neurologic and general well-being of patients from a tertiary care centre. J Indian Med Assoc. 2012;110:885–8.PubMed Mukhopadhyay P, Sinha U, Banerjee J, Choudhury AR, Philips CA. The effects in correction of anaemia in chronic kidney disease with erythropoietin therapy–preference to cardiovascular, neurologic and general well-being of patients from a tertiary care centre. J Indian Med Assoc. 2012;110:885–8.PubMed
23.
Zurück zum Zitat Covic A, Jackson J, Hadfield A, Pike J, Siriopol D. Real-world impact of cardiovascular disease and anemia on quality of life and productivity in patients with non-dialysis-dependent chronic kidney disease. Adv Ther. 2017;34:1662–72.PubMedCrossRefPubMedCentral Covic A, Jackson J, Hadfield A, Pike J, Siriopol D. Real-world impact of cardiovascular disease and anemia on quality of life and productivity in patients with non-dialysis-dependent chronic kidney disease. Adv Ther. 2017;34:1662–72.PubMedCrossRefPubMedCentral
24.
Zurück zum Zitat Finkelstein FO, Wuerth D, Finkelstein SH. Health related quality of life and the CKD patient: challenges for the nephrology community. Kidney Int. 2009;76:946–52.PubMedCrossRef Finkelstein FO, Wuerth D, Finkelstein SH. Health related quality of life and the CKD patient: challenges for the nephrology community. Kidney Int. 2009;76:946–52.PubMedCrossRef
25.
Zurück zum Zitat Rao M, Pereira BJ. Optimal anemia management reduces cardiovascular morbidity, mortality, and costs in chronic kidney disease. Kidney Int. 2005;68:1432–8.PubMedCrossRef Rao M, Pereira BJ. Optimal anemia management reduces cardiovascular morbidity, mortality, and costs in chronic kidney disease. Kidney Int. 2005;68:1432–8.PubMedCrossRef
26.
Zurück zum Zitat Garlo K, Williams D, Lucas L, et al. Severity of anemia predicts hospital length of stay but not readmission in patients with chronic kidney disease: a retrospective cohort study. Medicine (Baltimore). 2015;94:e964.CrossRef Garlo K, Williams D, Lucas L, et al. Severity of anemia predicts hospital length of stay but not readmission in patients with chronic kidney disease: a retrospective cohort study. Medicine (Baltimore). 2015;94:e964.CrossRef
27.
Zurück zum Zitat Maddux FW, Shetty S, del Aguila MA, Nelson MA, Murray BM. Effect of erythropoiesis-stimulating agents on healthcare utilization, costs, and outcomes in chronic kidney disease. Ann Pharmacother. 2007;41:1761–9.PubMedCrossRef Maddux FW, Shetty S, del Aguila MA, Nelson MA, Murray BM. Effect of erythropoiesis-stimulating agents on healthcare utilization, costs, and outcomes in chronic kidney disease. Ann Pharmacother. 2007;41:1761–9.PubMedCrossRef
28.
Zurück zum Zitat Nissenson AR, Wade S, Goodnough T, Knight K, Dubois RW. Economic burden of anemia in an insured population. J Manag Care Pharm. 2005;11:565–74.PubMedCrossRef Nissenson AR, Wade S, Goodnough T, Knight K, Dubois RW. Economic burden of anemia in an insured population. J Manag Care Pharm. 2005;11:565–74.PubMedCrossRef
29.
Zurück zum Zitat Arantes LH Jr, Crawford J, Gascon P, et al. A quick scoping review of efficacy, safety, economic, and health-related quality-of-life outcomes of short- and long-acting erythropoiesis-stimulating agents in the treatment of chemotherapy-induced anemia and chronic kidney disease anemia. Crit Rev Oncol Hematol. 2018;129:79–90.PubMedCrossRef Arantes LH Jr, Crawford J, Gascon P, et al. A quick scoping review of efficacy, safety, economic, and health-related quality-of-life outcomes of short- and long-acting erythropoiesis-stimulating agents in the treatment of chemotherapy-induced anemia and chronic kidney disease anemia. Crit Rev Oncol Hematol. 2018;129:79–90.PubMedCrossRef
30.
Zurück zum Zitat Locatelli F, Pisoni RL, Combe C, et al. Anaemia in haemodialysis patients of five European countries: association with morbidity and mortality in the Dialysis Outcomes and Practice Patterns Study (DOPPS). Nephrol Dial Transpl. 2004;19:121–32.CrossRef Locatelli F, Pisoni RL, Combe C, et al. Anaemia in haemodialysis patients of five European countries: association with morbidity and mortality in the Dialysis Outcomes and Practice Patterns Study (DOPPS). Nephrol Dial Transpl. 2004;19:121–32.CrossRef
31.
Zurück zum Zitat Bennett WM. A multicenter clinical trial of epoetin beta for anemia of end-stage renal disease. J Am Soc Nephrol. 1991;1:990–8.PubMed Bennett WM. A multicenter clinical trial of epoetin beta for anemia of end-stage renal disease. J Am Soc Nephrol. 1991;1:990–8.PubMed
32.
Zurück zum Zitat Rosati A, Ravaglia F, Panichi V. Improving erythropoiesis stimulating agent hyporesponsiveness in hemodialysis patients: the role of hepcidin and hemodiafiltration online. Blood Purif. 2018;45:139–46.PubMedCrossRef Rosati A, Ravaglia F, Panichi V. Improving erythropoiesis stimulating agent hyporesponsiveness in hemodialysis patients: the role of hepcidin and hemodiafiltration online. Blood Purif. 2018;45:139–46.PubMedCrossRef
33.
Zurück zum Zitat Sinha SD, Bandi VK, Bheemareddy BR, et al. Efficacy, tolerability and safety of darbepoetin alfa injection for the treatment of anemia associated with chronic kidney disease (CKD) undergoing dialysis: a randomized, phase-III trial. BMC Nephrol. 2019;20:90.PubMedCrossRefPubMedCentral Sinha SD, Bandi VK, Bheemareddy BR, et al. Efficacy, tolerability and safety of darbepoetin alfa injection for the treatment of anemia associated with chronic kidney disease (CKD) undergoing dialysis: a randomized, phase-III trial. BMC Nephrol. 2019;20:90.PubMedCrossRefPubMedCentral
34.
Zurück zum Zitat Woodland AL, Murphy SW, Curtis BM, Barrett BJ. Costs associated with intravenous darbepoetin versus epoetin therapy in hemodialysis patients: a randomized controlled trial. Can J Kidney Health Dis. 2017;4:2054358117716461.PubMedCrossRefPubMedCentral Woodland AL, Murphy SW, Curtis BM, Barrett BJ. Costs associated with intravenous darbepoetin versus epoetin therapy in hemodialysis patients: a randomized controlled trial. Can J Kidney Health Dis. 2017;4:2054358117716461.PubMedCrossRefPubMedCentral
35.
Zurück zum Zitat Chen N, Hao C, Liu BC, et al. Roxadustat treatment for anemia in patients undergoing long-term dialysis. N Engl J Med. 2019;381:1011–22.PubMedCrossRef Chen N, Hao C, Liu BC, et al. Roxadustat treatment for anemia in patients undergoing long-term dialysis. N Engl J Med. 2019;381:1011–22.PubMedCrossRef
36.
Zurück zum Zitat 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;69:815–26.PubMedCrossRef 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;69:815–26.PubMedCrossRef
37.
Zurück zum Zitat Kaplan JM, Sharma N, Dikdan S. Hypoxia-inducible factor and its role in the management of anemia in chronic kidney disease. Int J Mol Sci. 2018;19:E389.PubMedCrossRef Kaplan JM, Sharma N, Dikdan S. Hypoxia-inducible factor and its role in the management of anemia in chronic kidney disease. Int J Mol Sci. 2018;19:E389.PubMedCrossRef
38.
Zurück zum Zitat Sakaguchi Y, Hamano T, Wada A, Masakane I. Types of erythropoietin-stimulating agents and mortality among patients undergoing hemodialysis. J Am Soc Nephrol. 2019;30:1037–48.PubMedCrossRefPubMedCentral Sakaguchi Y, Hamano T, Wada A, Masakane I. Types of erythropoietin-stimulating agents and mortality among patients undergoing hemodialysis. J Am Soc Nephrol. 2019;30:1037–48.PubMedCrossRefPubMedCentral
39.
Zurück zum Zitat Macdougall IC, Hutton RD, Cavill I, Coles GA, Williams JD. Poor response to treatment of renal anaemia with erythropoietin corrected by iron given intravenously. BMJ. 1989;299:157–8.PubMedCrossRefPubMedCentral Macdougall IC, Hutton RD, Cavill I, Coles GA, Williams JD. Poor response to treatment of renal anaemia with erythropoietin corrected by iron given intravenously. BMJ. 1989;299:157–8.PubMedCrossRefPubMedCentral
41.
42.
Zurück zum Zitat Macdougall IC, Bircher AJ, Eckardt KU, et al. Iron management in chronic kidney disease: conclusions from a "Kidney Disease: Improving Global Outcomes" (KDIGO) Controversies Conference. Kidney Int. 2016;89:28–39.PubMedCrossRef Macdougall IC, Bircher AJ, Eckardt KU, et al. Iron management in chronic kidney disease: conclusions from a "Kidney Disease: Improving Global Outcomes" (KDIGO) Controversies Conference. Kidney Int. 2016;89:28–39.PubMedCrossRef
43.
44.
Zurück zum Zitat Macdougall IC, Vernon K. Complement activation-related pseudo-allergy: a fresh look at hypersensitivity reactions to intravenous iron. Am J Nephrol. 2017;45:60–2.PubMedCrossRef Macdougall IC, Vernon K. Complement activation-related pseudo-allergy: a fresh look at hypersensitivity reactions to intravenous iron. Am J Nephrol. 2017;45:60–2.PubMedCrossRef
45.
Zurück zum Zitat Wang C, Graham DJ, Kane RC, et al. Comparative risk of anaphylactic reactions associated with intravenous iron products. JAMA. 2015;314:2062–8.PubMedCrossRef Wang C, Graham DJ, Kane RC, et al. Comparative risk of anaphylactic reactions associated with intravenous iron products. JAMA. 2015;314:2062–8.PubMedCrossRef
46.
Zurück zum Zitat Roger SD. Practical considerations for iron therapy in the management of anaemia in patients with chronic kidney disease. Clin Kidney J. 2017;10:i9–i15.PubMedCrossRefPubMedCentral Roger SD. Practical considerations for iron therapy in the management of anaemia in patients with chronic kidney disease. Clin Kidney J. 2017;10:i9–i15.PubMedCrossRefPubMedCentral
47.
Zurück zum Zitat Kaushik T, Yaqoob MM. Lessons learned from peginesatide in the treatment of anemia associated with chronic kidney disease in patients on dialysis. Biologics. 2013;7:243–6.PubMedPubMedCentral Kaushik T, Yaqoob MM. Lessons learned from peginesatide in the treatment of anemia associated with chronic kidney disease in patients on dialysis. Biologics. 2013;7:243–6.PubMedPubMedCentral
49.
Zurück zum Zitat Goodkin DA, Fuller DS, Robinson BM, et al. Naturally occurring higher hemoglobin concentration does not increase mortality among hemodialysis patients. J Am Soc Nephrol. 2011;22:358–65.PubMedCrossRefPubMedCentral Goodkin DA, Fuller DS, Robinson BM, et al. Naturally occurring higher hemoglobin concentration does not increase mortality among hemodialysis patients. J Am Soc Nephrol. 2011;22:358–65.PubMedCrossRefPubMedCentral
50.
Zurück zum Zitat Akaishi M, Hiroe M, Hada Y, et al. Effect of anemia correction on left ventricular hypertrophy in patients with modestly high hemoglobin level and chronic kidney disease. J Cardiol. 2013;62:249–56.PubMedCrossRef Akaishi M, Hiroe M, Hada Y, et al. Effect of anemia correction on left ventricular hypertrophy in patients with modestly high hemoglobin level and chronic kidney disease. J Cardiol. 2013;62:249–56.PubMedCrossRef
51.
Zurück zum Zitat Hayashi T, Uemura Y, Kumagai M, et al. Effect of achieved hemoglobin level on renal outcome in non-dialysis chronic kidney disease (CKD) patients receiving epoetin beta pegol: MIRcerA Clinical Evidence on Renal Survival in CKD patients with renal anemia (MIRACLE-CKD Study). Clin Exp Nephrol. 2019;23:349–61.PubMedCrossRef Hayashi T, Uemura Y, Kumagai M, et al. Effect of achieved hemoglobin level on renal outcome in non-dialysis chronic kidney disease (CKD) patients receiving epoetin beta pegol: MIRcerA Clinical Evidence on Renal Survival in CKD patients with renal anemia (MIRACLE-CKD Study). Clin Exp Nephrol. 2019;23:349–61.PubMedCrossRef
52.
Zurück zum Zitat Ritz E, Laville M, Bilous RW, et al. Target level for hemoglobin correction in patients with diabetes and CKD: primary results of the Anemia Correction in Diabetes (ACORD) Study. Am J Kidney Dis. 2007;49:194–207.PubMedCrossRef Ritz E, Laville M, Bilous RW, et al. Target level for hemoglobin correction in patients with diabetes and CKD: primary results of the Anemia Correction in Diabetes (ACORD) Study. Am J Kidney Dis. 2007;49:194–207.PubMedCrossRef
53.
Zurück zum Zitat Drueke TB, Locatelli F, Clyne N, et al. Normalization of hemoglobin level in patients with chronic kidney disease and anemia. N Engl J Med. 2006;355:2071–84.PubMedCrossRef Drueke TB, Locatelli F, Clyne N, et al. Normalization of hemoglobin level in patients with chronic kidney disease and anemia. N Engl J Med. 2006;355:2071–84.PubMedCrossRef
54.
Zurück zum Zitat Solomon SD, Uno H, Lewis EF, et al. Erythropoietic response and outcomes in kidney disease and type 2 diabetes. N Engl J Med. 2010;363:1146–55.PubMedCrossRef Solomon SD, Uno H, Lewis EF, et al. Erythropoietic response and outcomes in kidney disease and type 2 diabetes. N Engl J Med. 2010;363:1146–55.PubMedCrossRef
55.
Zurück zum Zitat Park H, Liu X, Henry L, Harman J, Ross EA. Trends in anemia care in non-dialysis-dependent chronic kidney disease (CKD) patients in the United States (2006–2015). BMC Nephrol. 2018;19:318.PubMedCrossRefPubMedCentral Park H, Liu X, Henry L, Harman J, Ross EA. Trends in anemia care in non-dialysis-dependent chronic kidney disease (CKD) patients in the United States (2006–2015). BMC Nephrol. 2018;19:318.PubMedCrossRefPubMedCentral
56.
Zurück zum Zitat Li S, Guo H, Kabadi S, et al. Unexpected medical consequences of revised ESA label in non-dialysis-dependent chronic kidney disease patients with anemia. J Am Soc Nephrol. 2016;27:814A.CrossRef Li S, Guo H, Kabadi S, et al. Unexpected medical consequences of revised ESA label in non-dialysis-dependent chronic kidney disease patients with anemia. J Am Soc Nephrol. 2016;27:814A.CrossRef
57.
Zurück zum Zitat Collister D, Komenda P, Hiebert B, et al. The effect of erythropoietin-stimulating agents on health-related quality of life in anemia of chronic kidney disease: a systematic review and meta-analysis. Ann Intern Med. 2016;164:472–8.PubMedCrossRef Collister D, Komenda P, Hiebert B, et al. The effect of erythropoietin-stimulating agents on health-related quality of life in anemia of chronic kidney disease: a systematic review and meta-analysis. Ann Intern Med. 2016;164:472–8.PubMedCrossRef
58.
Zurück zum Zitat Spinowitz B, Pecoits-Filho R, Winkelmayer WC, et al. Economic and quality of life burden of anemia on patients with CKD on dialysis: a systematic review. J Med Econ. 2019;22:593–604.PubMedCrossRef Spinowitz B, Pecoits-Filho R, Winkelmayer WC, et al. Economic and quality of life burden of anemia on patients with CKD on dialysis: a systematic review. J Med Econ. 2019;22:593–604.PubMedCrossRef
59.
Zurück zum Zitat Johansen KL, Finkelstein FO, Revicki DA, et al. Systematic review of the impact of erythropoiesis-stimulating agents on fatigue in dialysis patients. Nephrol Dial Transpl. 2012;27:2418–25.CrossRef Johansen KL, Finkelstein FO, Revicki DA, et al. Systematic review of the impact of erythropoiesis-stimulating agents on fatigue in dialysis patients. Nephrol Dial Transpl. 2012;27:2418–25.CrossRef
60.
Zurück zum Zitat Muirhead N, Keown PA, Churchill DN, et al. Dialysis patients treated with Epoetin alpha show improved exercise tolerance and physical function: a new analysis of the Canadian Erythropoietin Study Group trial. Hemodial Int. 2011;15:87–94.PubMedCrossRef Muirhead N, Keown PA, Churchill DN, et al. Dialysis patients treated with Epoetin alpha show improved exercise tolerance and physical function: a new analysis of the Canadian Erythropoietin Study Group trial. Hemodial Int. 2011;15:87–94.PubMedCrossRef
61.
Zurück zum Zitat Fox KM, Yee J, Cong Z, et al. Transfusion burden in non-dialysis chronic kidney disease patients with persistent anemia treated in routine clinical practice: a retrospective observational study. BMC Nephrol. 2012;13:5.PubMedCrossRefPubMedCentral Fox KM, Yee J, Cong Z, et al. Transfusion burden in non-dialysis chronic kidney disease patients with persistent anemia treated in routine clinical practice: a retrospective observational study. BMC Nephrol. 2012;13:5.PubMedCrossRefPubMedCentral
62.
Zurück zum Zitat Bohlius J, Bohlke K, Castelli R, et al. Management of cancer-associated anemia with erythropoiesis-stimulating agents: ASCO/ASH clinical practice guideline update. Blood Adv. 2019;3:1197–210.PubMedCrossRefPubMedCentral Bohlius J, Bohlke K, Castelli R, et al. Management of cancer-associated anemia with erythropoiesis-stimulating agents: ASCO/ASH clinical practice guideline update. Blood Adv. 2019;3:1197–210.PubMedCrossRefPubMedCentral
63.
Zurück zum Zitat Lattanzio F, Corsonello A, Montesanto A, et al. Disentangling the impact of chronic kidney disease, anemia, and mobility limitation on mortality in older patients discharged from hospital. J Gerontol A Biol Sci Med Sci. 2015;70:1120–7.PubMedCrossRef Lattanzio F, Corsonello A, Montesanto A, et al. Disentangling the impact of chronic kidney disease, anemia, and mobility limitation on mortality in older patients discharged from hospital. J Gerontol A Biol Sci Med Sci. 2015;70:1120–7.PubMedCrossRef
64.
Zurück zum Zitat Kuragano T, Mizusaki K, Kimura T, Nakanishi T. Anemia management considering the pathophysiology of elderly chronic kidney disease patients. Contrib Nephrol. 2019;198:135–43.PubMedCrossRef Kuragano T, Mizusaki K, Kimura T, Nakanishi T. Anemia management considering the pathophysiology of elderly chronic kidney disease patients. Contrib Nephrol. 2019;198:135–43.PubMedCrossRef
65.
Zurück zum Zitat Dousdampanis P, Trigka K, Fourtounas C. Prevalence of anemia in patients with type II diabetes and mild to moderate chronic kidney disease and the impact of anti-RAS medications. Saudi J Kidney Dis Transpl. 2014;25:552–7.PubMedCrossRef Dousdampanis P, Trigka K, Fourtounas C. Prevalence of anemia in patients with type II diabetes and mild to moderate chronic kidney disease and the impact of anti-RAS medications. Saudi J Kidney Dis Transpl. 2014;25:552–7.PubMedCrossRef
66.
Zurück zum Zitat Pappa M, Dounousi E, Duni A, Katopodis K. Less known pathophysiological mechanisms of anemia in patients with diabetic nephropathy. Int Urol Nephrol. 2015;47:1365–72.PubMedCrossRef Pappa M, Dounousi E, Duni A, Katopodis K. Less known pathophysiological mechanisms of anemia in patients with diabetic nephropathy. Int Urol Nephrol. 2015;47:1365–72.PubMedCrossRef
67.
Zurück zum Zitat Fujita Y, Doi Y, Hamano T, et al. Low erythropoietin levels predict faster renal function decline in diabetic patients with anemia: a prospective cohort study. Sci Rep. 2019;9:14871.PubMedCrossRefPubMedCentral Fujita Y, Doi Y, Hamano T, et al. Low erythropoietin levels predict faster renal function decline in diabetic patients with anemia: a prospective cohort study. Sci Rep. 2019;9:14871.PubMedCrossRefPubMedCentral
68.
Zurück zum Zitat Stevens PE. Anaemia, diabetes and chronic kidney disease: where are we now? J Ren Care. 2012;38(Suppl 1):67–77.PubMedCrossRef Stevens PE. Anaemia, diabetes and chronic kidney disease: where are we now? J Ren Care. 2012;38(Suppl 1):67–77.PubMedCrossRef
70.
Zurück zum Zitat Winocour PH. Diabetes and chronic kidney disease: an increasingly common multi-morbid disease in need of a paradigm shift in care. Diabet Med. 2018;35:300–5.PubMedCrossRef Winocour PH. Diabetes and chronic kidney disease: an increasingly common multi-morbid disease in need of a paradigm shift in care. Diabet Med. 2018;35:300–5.PubMedCrossRef
71.
Zurück zum Zitat Fowler MJ. Microvascular and macrovascular complications of diabetes. Clinical Diabetes. 2008;26:77–82.CrossRef Fowler MJ. Microvascular and macrovascular complications of diabetes. Clinical Diabetes. 2008;26:77–82.CrossRef
72.
Zurück zum Zitat Bajaj S, Makkar BM, Abichandani VK, et al. Management of anemia in patients with diabetic kidney disease: a consensus statement. Indian J Endocrinol Metab. 2016;20:268–81.PubMedCrossRefPubMedCentral Bajaj S, Makkar BM, Abichandani VK, et al. Management of anemia in patients with diabetic kidney disease: a consensus statement. Indian J Endocrinol Metab. 2016;20:268–81.PubMedCrossRefPubMedCentral
73.
Zurück zum Zitat Portoles J, Gorriz JL, Rubio E, et al. The development of anemia is associated to poor prognosis in NKF/KDOQI stage 3 chronic kidney disease. BMC Nephrol. 2013;14:2.PubMedCrossRefPubMedCentral Portoles J, Gorriz JL, Rubio E, et al. The development of anemia is associated to poor prognosis in NKF/KDOQI stage 3 chronic kidney disease. BMC Nephrol. 2013;14:2.PubMedCrossRefPubMedCentral
74.
Zurück zum Zitat Moss AH, Davison SN. How the ESRD quality incentive program could potentially improve quality of life for patients on dialysis. Clin J Am Soc Nephrol. 2015;10:888–93.PubMedCrossRefPubMedCentral Moss AH, Davison SN. How the ESRD quality incentive program could potentially improve quality of life for patients on dialysis. Clin J Am Soc Nephrol. 2015;10:888–93.PubMedCrossRefPubMedCentral
75.
Zurück zum Zitat Valliant A, Hofmann RM. Managing dialysis patients who develop anemia caused by chronic kidney disease: focus on peginesatide. Int J Nanomedicine. 2013;8:3297–307.PubMedPubMedCentral Valliant A, Hofmann RM. Managing dialysis patients who develop anemia caused by chronic kidney disease: focus on peginesatide. Int J Nanomedicine. 2013;8:3297–307.PubMedPubMedCentral
76.
Zurück zum Zitat Rostoker G, Griuncelli M, Loridon C, et al. Hemodialysis-associated hemosiderosis in the era of erythropoiesis-stimulating agents: a MRI study. Am J Med. 2012;125(991–9):e1. Rostoker G, Griuncelli M, Loridon C, et al. Hemodialysis-associated hemosiderosis in the era of erythropoiesis-stimulating agents: a MRI study. Am J Med. 2012;125(991–9):e1.
77.
Zurück zum Zitat Besarab A, Bolton WK, Browne JK, et al. The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. N Engl J Med. 1998;339:584–90.PubMedCrossRef Besarab A, Bolton WK, Browne JK, et al. The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. N Engl J Med. 1998;339:584–90.PubMedCrossRef
78.
Zurück zum Zitat Gafter-Gvili A, Ayalon-Dangur I, Cooper L, et al. Posttransplantation anemia in kidney transplant recipients: a retrospective cohort study. Medicine (Baltimore). 2017;96:e7735.CrossRef Gafter-Gvili A, Ayalon-Dangur I, Cooper L, et al. Posttransplantation anemia in kidney transplant recipients: a retrospective cohort study. Medicine (Baltimore). 2017;96:e7735.CrossRef
79.
Zurück zum Zitat Gurlek Demirci B, Sezer S, Sayin CB, et al. Post-transplantation anemia predicts cardiovascular morbidity and poor graft function in kidney transplant recipients. Transpl Proc. 2015;47:1178–81.CrossRef Gurlek Demirci B, Sezer S, Sayin CB, et al. Post-transplantation anemia predicts cardiovascular morbidity and poor graft function in kidney transplant recipients. Transpl Proc. 2015;47:1178–81.CrossRef
80.
Zurück zum Zitat Tsujita M, Kosugi T, Goto N, et al. The effect of maintaining high hemoglobin levels on long-term kidney function in kidney transplant recipients: a randomized controlled trial. Nephrol Dial Transpl. 2019;34:1409–16.CrossRef Tsujita M, Kosugi T, Goto N, et al. The effect of maintaining high hemoglobin levels on long-term kidney function in kidney transplant recipients: a randomized controlled trial. Nephrol Dial Transpl. 2019;34:1409–16.CrossRef
81.
Zurück zum Zitat Kitamura K, Nakai K, Fujii H, Ishimura T, Fujisawa M, Nishi S. Pre-transplant erythropoiesis-stimulating agent hypo-responsiveness and post-transplant anemia. Transpl Proc. 2015;47:1820–4.CrossRef Kitamura K, Nakai K, Fujii H, Ishimura T, Fujisawa M, Nishi S. Pre-transplant erythropoiesis-stimulating agent hypo-responsiveness and post-transplant anemia. Transpl Proc. 2015;47:1820–4.CrossRef
82.
84.
Zurück zum Zitat Kato S, Ochiai N, Takano H, et al. TP0463518, a novel prolyl hydroxylase inhibitor, specifically induces erythropoietin production in the liver. J Pharmacol Exp Ther. 2019;371:675–83.PubMedCrossRef Kato S, Ochiai N, Takano H, et al. TP0463518, a novel prolyl hydroxylase inhibitor, specifically induces erythropoietin production in the liver. J Pharmacol Exp Ther. 2019;371:675–83.PubMedCrossRef
85.
Zurück zum Zitat Shah YM, Matsubara T, Ito S, Yim SH, Gonzalez FJ. Intestinal hypoxia-inducible transcription factors are essential for iron absorption following iron deficiency. Cell Metab. 2009;9:152–64.PubMedCrossRefPubMedCentral Shah YM, Matsubara T, Ito S, Yim SH, Gonzalez FJ. Intestinal hypoxia-inducible transcription factors are essential for iron absorption following iron deficiency. Cell Metab. 2009;9:152–64.PubMedCrossRefPubMedCentral
86.
Zurück zum Zitat Chen N, Qian J, Chen J, et al. Phase 2 studies of oral hypoxia-inducible factor prolyl hydroxylase inhibitor FG-4592 for treatment of anemia in China. Nephrol Dial Transpl. 2017;32:1373–86.CrossRef Chen N, Qian J, Chen J, et al. Phase 2 studies of oral hypoxia-inducible factor prolyl hydroxylase inhibitor FG-4592 for treatment of anemia in China. Nephrol Dial Transpl. 2017;32:1373–86.CrossRef
87.
Zurück zum Zitat Provenzano R, Besarab A, Wright S, 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;67:912–24.PubMedCrossRef Provenzano R, Besarab A, Wright S, 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;67:912–24.PubMedCrossRef
88.
Zurück zum Zitat Besarab A, Chernyavskaya E, Motylev I, et al. Roxadustat (FG-4592): correction of anemia in incident dialysis patients. J Am Soc Nephrol. 2016;27:1225–333.PubMedCrossRef Besarab A, Chernyavskaya E, Motylev I, et al. Roxadustat (FG-4592): correction of anemia in incident dialysis patients. J Am Soc Nephrol. 2016;27:1225–333.PubMedCrossRef
90.
Zurück zum Zitat Charytan C, Manllo-Karim R, Martin ER, et al. SIERRAS: a phase 3, open-label, randomized, active-controlled study of the efficacy and safety of roxadustat in the maintenance treatment of anemia in subjects with ESRD on stable dialysis [abstract SA-PO227]. J Am Soc Nephrol. 2019;30:822. Charytan C, Manllo-Karim R, Martin ER, et al. SIERRAS: a phase 3, open-label, randomized, active-controlled study of the efficacy and safety of roxadustat in the maintenance treatment of anemia in subjects with ESRD on stable dialysis [abstract SA-PO227]. J Am Soc Nephrol. 2019;30:822.
91.
Zurück zum Zitat Fishbane S, Pollock CA, El-Shahawy MA, et al. ROCKIES: an international, phase 3, randomized, open-label, active-controlled study of roxadustat for anemia in dialysis-dependent CKD patients [abstract TH-OR022]. J Am Soc Nephrol. 2019;30:6.CrossRef Fishbane S, Pollock CA, El-Shahawy MA, et al. ROCKIES: an international, phase 3, randomized, open-label, active-controlled study of roxadustat for anemia in dialysis-dependent CKD patients [abstract TH-OR022]. J Am Soc Nephrol. 2019;30:6.CrossRef
92.
Zurück zum Zitat Esposito C, Csiky B, Tataradze A, Reusch M, Han C, Sulowicz W. Two phase 3, multicenter, randomized studies of intermittent oral roxadustat in anemic CKD patients on (PYRENEES) and not on (ALPS) dialysis [abstract SA-PO225]. J Am Soc Nephrol. 2019;30:822. Esposito C, Csiky B, Tataradze A, Reusch M, Han C, Sulowicz W. Two phase 3, multicenter, randomized studies of intermittent oral roxadustat in anemic CKD patients on (PYRENEES) and not on (ALPS) dialysis [abstract SA-PO225]. J Am Soc Nephrol. 2019;30:822.
93.
Zurück zum Zitat Provenzano R, Evgeny S, Liubov E, et al. HIMALAYAS: a phase 3, randomized, open-label, active-controlled study of the efficacy and safety of roxadustat in the treatment of anemia in incident-dialysis patients [abstract TH-OR021]. J Am Soc Nephrol. 2019;30:5. Provenzano R, Evgeny S, Liubov E, et al. HIMALAYAS: a phase 3, randomized, open-label, active-controlled study of the efficacy and safety of roxadustat in the treatment of anemia in incident-dialysis patients [abstract TH-OR021]. J Am Soc Nephrol. 2019;30:5.
94.
Zurück zum Zitat Provenzano R, Besarab A, Sun CH, 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;11:982–91.PubMedCrossRefPubMedCentral Provenzano R, Besarab A, Sun CH, 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;11:982–91.PubMedCrossRefPubMedCentral
95.
Zurück zum Zitat Besarab A, Provenzano R, Hertel J, 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 Transpl. 2015;30:1665–733.CrossRef Besarab A, Provenzano R, Hertel J, 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 Transpl. 2015;30:1665–733.CrossRef
96.
Zurück zum Zitat Chen N, Hao C, Peng X, et al. Roxadustat for anemia in patients with kidney disease not receiving dialysis. N Engl J Med. 2019;381:1001–100.PubMedCrossRef Chen N, Hao C, Peng X, et al. Roxadustat for anemia in patients with kidney disease not receiving dialysis. N Engl J Med. 2019;381:1001–100.PubMedCrossRef
97.
Zurück zum Zitat Coyne DW, Roger SD, Shin SK, et al. ANDES: a phase 3, randomized, double-blind, placebo controlled study of the efficacy and safety of roxadustat for the treatment of anemia in CKD patients not on dialysis [abstract SA-PO228]. J Am Soc Nephrol. 2019;30:822–3. Coyne DW, Roger SD, Shin SK, et al. ANDES: a phase 3, randomized, double-blind, placebo controlled study of the efficacy and safety of roxadustat for the treatment of anemia in CKD patients not on dialysis [abstract SA-PO228]. J Am Soc Nephrol. 2019;30:822–3.
98.
Zurück zum Zitat Fishbane S, El-Shahawy MA, Pecoits-Filho R, et al. OLYMPUS: a phase 3, randomized, double-blind, placebo-controlled, international study of roxadustat efficacy in patients with non-dialysis-dependent (NDD) CKD and anemia [abstract TH-OR023]. J Am Soc Nephrol. 2019;30:6.CrossRef Fishbane S, El-Shahawy MA, Pecoits-Filho R, et al. OLYMPUS: a phase 3, randomized, double-blind, placebo-controlled, international study of roxadustat efficacy in patients with non-dialysis-dependent (NDD) CKD and anemia [abstract TH-OR023]. J Am Soc Nephrol. 2019;30:6.CrossRef
99.
Zurück zum Zitat Akizawa T, Tsubakihara Y, Nangaku M, et al. Effects of daprodustat, a novel hypoxia-inducible factor prolyl hydroxylase inhibitor on anemia management in japanese hemodialysis subjects. Am J Nephrol. 2017;45:127–35.PubMedCrossRef Akizawa T, Tsubakihara Y, Nangaku M, et al. Effects of daprodustat, a novel hypoxia-inducible factor prolyl hydroxylase inhibitor on anemia management in japanese hemodialysis subjects. Am J Nephrol. 2017;45:127–35.PubMedCrossRef
100.
Zurück zum Zitat Holdstock L, Meadowcroft AM, Maier R, et al. Four-week studies of oral hypoxia-inducible factor-prolyl hydroxylase inhibitor GSK1278863 for treatment of anemia. J Am Soc Nephrol. 2016;27:1234–44.PubMedCrossRef Holdstock L, Meadowcroft AM, Maier R, et al. Four-week studies of oral hypoxia-inducible factor-prolyl hydroxylase inhibitor GSK1278863 for treatment of anemia. J Am Soc Nephrol. 2016;27:1234–44.PubMedCrossRef
101.
Zurück zum Zitat Cizman B, Sykes AP, Paul G, Zeig S, Cobitz AR. An exploratory study of daprodustat in erythropoietin-hyporesponsive subjects. Kidney Int Rep. 2018;3:841–50.PubMedCrossRefPubMedCentral Cizman B, Sykes AP, Paul G, Zeig S, Cobitz AR. An exploratory study of daprodustat in erythropoietin-hyporesponsive subjects. Kidney Int Rep. 2018;3:841–50.PubMedCrossRefPubMedCentral
102.
Zurück zum Zitat Meadowcroft AM, Cizman B, Holdstock L, et al. Daprodustat for anemia: a 24-week, open-label, randomized controlled trial in participants on hemodialysis. Clin Kidney J. 2019;12:139–48.PubMedCrossRef Meadowcroft AM, Cizman B, Holdstock L, et al. Daprodustat for anemia: a 24-week, open-label, randomized controlled trial in participants on hemodialysis. Clin Kidney J. 2019;12:139–48.PubMedCrossRef
103.
Zurück zum Zitat Tsubakihara Y, Akizawa T, Nangaku M, et al. A 24-week anemia correction study of daprodustat in japanese dialysis patients. Ther Apher Dial. 2020;24:108–14.PubMedCrossRef Tsubakihara Y, Akizawa T, Nangaku M, et al. A 24-week anemia correction study of daprodustat in japanese dialysis patients. Ther Apher Dial. 2020;24:108–14.PubMedCrossRef
104.
Zurück zum Zitat Holdstock L, Cizman B, Meadowcroft AM, et al. Daprodustat for anemia: a 24-week, open-label, randomized controlled trial in participants with chronic kidney disease. Clin Kidney J. 2019;12:129–38.PubMedCrossRef Holdstock L, Cizman B, Meadowcroft AM, et al. Daprodustat for anemia: a 24-week, open-label, randomized controlled trial in participants with chronic kidney disease. Clin Kidney J. 2019;12:129–38.PubMedCrossRef
105.
Zurück zum Zitat Brigandi RA, Johnson B, Oei C, et al. A novel hypoxia-inducible factor-prolyl hydroxylase inhibitor (GSK1278863) for anemia in CKD: a 28-day, phase 2A randomized trial. Am J Kidney Dis. 2016;67:861–71.PubMedCrossRef Brigandi RA, Johnson B, Oei C, et al. A novel hypoxia-inducible factor-prolyl hydroxylase inhibitor (GSK1278863) for anemia in CKD: a 28-day, phase 2A randomized trial. Am J Kidney Dis. 2016;67:861–71.PubMedCrossRef
106.
Zurück zum Zitat Haase VH, Chertow GM, Block GA, et al. Effects of vadadustat on hemoglobin concentrations in patients receiving hemodialysis previously treated with erythropoiesis-stimulating agents. Nephrol Dial Transpl. 2019;34:90–9.CrossRef Haase VH, Chertow GM, Block GA, et al. Effects of vadadustat on hemoglobin concentrations in patients receiving hemodialysis previously treated with erythropoiesis-stimulating agents. Nephrol Dial Transpl. 2019;34:90–9.CrossRef
107.
Zurück zum Zitat Pergola PE, Spinowitz BS, Hartman CS, Maroni BJ, Haase VH. Vadadustat, a novel oral HIF stabilizer, provides effective anemia treatment in nondialysis-dependent chronic kidney disease. Kidney Int. 2016;90:1115–22.PubMedCrossRef Pergola PE, Spinowitz BS, Hartman CS, Maroni BJ, Haase VH. Vadadustat, a novel oral HIF stabilizer, provides effective anemia treatment in nondialysis-dependent chronic kidney disease. Kidney Int. 2016;90:1115–22.PubMedCrossRef
108.
Zurück zum Zitat Martin ER, Smith MT, Maroni BJ, Zuraw QC, deGoma EM. Clinical trial of vadadustat in patients with anemia secondary to stage 3 or 4 chronic kidney disease. Am J Nephrol. 2017;45:380–8.PubMedCrossRef Martin ER, Smith MT, Maroni BJ, Zuraw QC, deGoma EM. Clinical trial of vadadustat in patients with anemia secondary to stage 3 or 4 chronic kidney disease. Am J Nephrol. 2017;45:380–8.PubMedCrossRef
109.
Zurück zum Zitat Macdougall IC, Akizawa T, Berns JS, Bernhardt T, Krueger T. Effects of Molidustat in the Treatment of Anemia in CKD. Clin J Am Soc Nephrol. 2019;14:28–39.PubMedCrossRef Macdougall IC, Akizawa T, Berns JS, Bernhardt T, Krueger T. Effects of Molidustat in the Treatment of Anemia in CKD. Clin J Am Soc Nephrol. 2019;14:28–39.PubMedCrossRef
110.
Zurück zum Zitat Akizawa T, Macdougall IC, Berns JS, et al. Long-term efficacy and safety of molidustat for anemia in chronic kidney disease: DIALOGUE extension studies. Am J Nephrol. 2019;49:271–80.PubMedCrossRef Akizawa T, Macdougall IC, Berns JS, et al. Long-term efficacy and safety of molidustat for anemia in chronic kidney disease: DIALOGUE extension studies. Am J Nephrol. 2019;49:271–80.PubMedCrossRef
111.
Zurück zum Zitat Akizawa T, Nangaku M, Yamaguchi T, et al. Enarodustat, conversion and maintenance therapy for anemia in hemodialysis patients: a randomized, placebo-controlled phase 2b trial followed by long-term trial. Nephron. 2019;143:77–85.PubMedCrossRef Akizawa T, Nangaku M, Yamaguchi T, et al. Enarodustat, conversion and maintenance therapy for anemia in hemodialysis patients: a randomized, placebo-controlled phase 2b trial followed by long-term trial. Nephron. 2019;143:77–85.PubMedCrossRef
112.
Zurück zum Zitat Akizawa T, Nangaku M, Yamaguchi T, et al. A placebo-controlled, randomized trial of enarodustat in patients with chronic kidney disease followed by long-term trial. Am J Nephrol. 2019;49:165–74.PubMedCrossRef Akizawa T, Nangaku M, Yamaguchi T, et al. A placebo-controlled, randomized trial of enarodustat in patients with chronic kidney disease followed by long-term trial. Am J Nephrol. 2019;49:165–74.PubMedCrossRef
113.
Zurück zum Zitat Parmar DV, Kansagra KA, Patel JC, et al. Outcomes of desidustat treatment in people with anemia and chronic kidney disease: a phase 2 study. Am J Nephrol. 2019;49:470–8.PubMedCrossRef Parmar DV, Kansagra KA, Patel JC, et al. Outcomes of desidustat treatment in people with anemia and chronic kidney disease: a phase 2 study. Am J Nephrol. 2019;49:470–8.PubMedCrossRef
114.
Zurück zum Zitat Shibata T, Nomura Y, Takada A, Aoki S, Katashima M, Murakami H. Evaluation of the effect of lanthanum carbonate hydrate on the pharmacokinetics of roxadustat in non-elderly healthy adult male subjects. J Clin Pharm Ther. 2018;43:633–9.PubMedCrossRef Shibata T, Nomura Y, Takada A, Aoki S, Katashima M, Murakami H. Evaluation of the effect of lanthanum carbonate hydrate on the pharmacokinetics of roxadustat in non-elderly healthy adult male subjects. J Clin Pharm Ther. 2018;43:633–9.PubMedCrossRef
115.
Zurück zum Zitat Shibata T, Nomura Y, Takada A, et al. Evaluation of food and spherical carbon adsorbent effects on the pharmacokinetics of roxadustat in healthy nonelderly adult male Japanese subjects. Clin Pharmacol Drug Dev. 2019;8:304–13.PubMedCrossRef Shibata T, Nomura Y, Takada A, et al. Evaluation of food and spherical carbon adsorbent effects on the pharmacokinetics of roxadustat in healthy nonelderly adult male Japanese subjects. Clin Pharmacol Drug Dev. 2019;8:304–13.PubMedCrossRef
116.
Zurück zum Zitat Groenendaal-van de Meent D, Adel MD, et al. Effect of moderate hepatic impairment on the pharmacokinetics and pharmacodynamics of roxadustat, an oral hypoxia-inducible factor prolyl hydroxylase inhibitor. Clin Drug Investig. 2016;36:743–51.PubMedCrossRefPubMedCentral Groenendaal-van de Meent D, Adel MD, et al. Effect of moderate hepatic impairment on the pharmacokinetics and pharmacodynamics of roxadustat, an oral hypoxia-inducible factor prolyl hydroxylase inhibitor. Clin Drug Investig. 2016;36:743–51.PubMedCrossRefPubMedCentral
117.
Zurück zum Zitat Hara K, Takahashi N, Wakamatsu A, Caltabiano S. Pharmacokinetics, pharmacodynamics and safety of single, oral doses of GSK1278863, a novel HIF-prolyl hydroxylase inhibitor, in healthy Japanese and Caucasian subjects. Drug Metab Pharmacokinet. 2015;30:410–8.PubMedCrossRef Hara K, Takahashi N, Wakamatsu A, Caltabiano S. Pharmacokinetics, pharmacodynamics and safety of single, oral doses of GSK1278863, a novel HIF-prolyl hydroxylase inhibitor, in healthy Japanese and Caucasian subjects. Drug Metab Pharmacokinet. 2015;30:410–8.PubMedCrossRef
118.
Zurück zum Zitat Buch A, Maroni BJ, Hartman CS. Dose exposure relationship of AKB-6548 is independent of the level of renal function. J Am Soc Nephrol. 2015;26:747A. Buch A, Maroni BJ, Hartman CS. Dose exposure relationship of AKB-6548 is independent of the level of renal function. J Am Soc Nephrol. 2015;26:747A.
119.
Zurück zum Zitat Bottcher M, Lentini S, Arens ER, et al. First-in-man-proof of concept study with molidustat: a novel selective oral HIF-prolyl hydroxylase inhibitor for the treatment of renal anaemia. Br J Clin Pharmacol. 2018;84:1557–655.PubMedCrossRefPubMedCentral Bottcher M, Lentini S, Arens ER, et al. First-in-man-proof of concept study with molidustat: a novel selective oral HIF-prolyl hydroxylase inhibitor for the treatment of renal anaemia. Br J Clin Pharmacol. 2018;84:1557–655.PubMedCrossRefPubMedCentral
120.
Zurück zum Zitat Kansagra KA, Parmar D, Jani RH, et al. Phase I clinical study of ZYAN1, a novel prolyl-hydroxylase (PHD) inhibitor to evaluate the safety, tolerability, and pharmacokinetics following oral administration in healthy volunteers. Clin Pharmacokinet. 2018;57:87–102.PubMedCrossRef Kansagra KA, Parmar D, Jani RH, et al. Phase I clinical study of ZYAN1, a novel prolyl-hydroxylase (PHD) inhibitor to evaluate the safety, tolerability, and pharmacokinetics following oral administration in healthy volunteers. Clin Pharmacokinet. 2018;57:87–102.PubMedCrossRef
123.
Zurück zum Zitat Akebia Therapeutics. Akebia Therapeutics announces approval of vadadustat in Japan for the treatment of anemia due to chronic kidney disease in dialysis-dependent and non-dialysis dependent adult patients. 2020. https://ir.akebia.com/press-releases. Accessed 23 July 2020. Akebia Therapeutics. Akebia Therapeutics announces approval of vadadustat in Japan for the treatment of anemia due to chronic kidney disease in dialysis-dependent and non-dialysis dependent adult patients. 2020. https://​ir.​akebia.​com/​press-releases. Accessed 23 July 2020.
125.
Zurück zum Zitat Barratt J, Andrić B, Tataradze A, et al. Roxadustat for the treatment of anaemia in chronic kidney disease patients not on dialysis: a phase 3, randomised, open-label, active-controlled study [abstract MO001]. Nephrol Dial Transpl. 2020;35:101–2. Barratt J, Andrić B, Tataradze A, et al. Roxadustat for the treatment of anaemia in chronic kidney disease patients not on dialysis: a phase 3, randomised, open-label, active-controlled study [abstract MO001]. Nephrol Dial Transpl. 2020;35:101–2.
126.
Zurück zum Zitat Nangaku M, Kondo K, Kokado Y, et al. Randomized, open-label, active-controlled (darbepoetin alfa), phase 3 study of vadadustat for treating anemia in non-dialysis-dependent CKD patients in Japan [abstract SA-PO229]. J Am Soc Nephrol. 2019;30:823. Nangaku M, Kondo K, Kokado Y, et al. Randomized, open-label, active-controlled (darbepoetin alfa), phase 3 study of vadadustat for treating anemia in non-dialysis-dependent CKD patients in Japan [abstract SA-PO229]. J Am Soc Nephrol. 2019;30:823.
127.
Zurück zum Zitat Nangaku M, Kondo K, Ueta K, et al. Randomized, double-blinded, active-controlled (darbepoetin alfa), phase 3 study of vadadustat in CKD patients with anemia on hemodialysis in Japan [abstract TH-OR024]. J Am Soc Nephrol. 2019;30:6. Nangaku M, Kondo K, Ueta K, et al. Randomized, double-blinded, active-controlled (darbepoetin alfa), phase 3 study of vadadustat in CKD patients with anemia on hemodialysis in Japan [abstract TH-OR024]. J Am Soc Nephrol. 2019;30:6.
130.
Zurück zum Zitat Provenzano R, Fishbane S, Wei L-J, et al. Pooled efficacy and cardiovascular (CV) analyses of roxadustat in the treatment of anemia in CKD patients on and not on dialysis [abstract FR-OR131]. J Am Soc Nephrol. 2019;30:B1.CrossRef Provenzano R, Fishbane S, Wei L-J, et al. Pooled efficacy and cardiovascular (CV) analyses of roxadustat in the treatment of anemia in CKD patients on and not on dialysis [abstract FR-OR131]. J Am Soc Nephrol. 2019;30:B1.CrossRef
131.
Zurück zum Zitat Beck J, Henschel C, Chou J, Lin A, Del Balzo U. Evaluation of the carcinogenic potential of Roxadustat (FG-4592), a small molecule inhibitor of hypoxia-inducible factor prolyl hydroxylase in CD-1 mice and Sprague Dawley rats. Int J Toxicol. 2017;36:427–39.PubMedCrossRef Beck J, Henschel C, Chou J, Lin A, Del Balzo U. Evaluation of the carcinogenic potential of Roxadustat (FG-4592), a small molecule inhibitor of hypoxia-inducible factor prolyl hydroxylase in CD-1 mice and Sprague Dawley rats. Int J Toxicol. 2017;36:427–39.PubMedCrossRef
132.
Zurück zum Zitat Seeley TW, Sternlicht MD, Klaus SJ, Neff TB, Liu DY. Induction of erythropoiesis by hypoxia-inducible factor prolyl hydroxylase inhibitors without promotion of tumor initiation, progression, or metastasis in a VEGF-sensitive model of spontaneous breast cancer. Hypoxia. 2017;5:1–9.PubMedCrossRefPubMedCentral Seeley TW, Sternlicht MD, Klaus SJ, Neff TB, Liu DY. Induction of erythropoiesis by hypoxia-inducible factor prolyl hydroxylase inhibitors without promotion of tumor initiation, progression, or metastasis in a VEGF-sensitive model of spontaneous breast cancer. Hypoxia. 2017;5:1–9.PubMedCrossRefPubMedCentral
134.
Zurück zum Zitat Hu Z, Tao H, Shi A, Pan J. The efficacy and economic evaluation of roxadustat treatment for anemia in patients with kidney disease not receiving dialysis. Expert Rev Pharmacoecon Outcomes Res. 2020;20:411–8.PubMedCrossRef Hu Z, Tao H, Shi A, Pan J. The efficacy and economic evaluation of roxadustat treatment for anemia in patients with kidney disease not receiving dialysis. Expert Rev Pharmacoecon Outcomes Res. 2020;20:411–8.PubMedCrossRef
135.
Zurück zum Zitat Johnson DW, Pollock CA, Macdougall IC. Erythropoiesis-stimulating agent hyporesponsiveness. Nephrology. 2007;12:321–30.PubMedCrossRef Johnson DW, Pollock CA, Macdougall IC. Erythropoiesis-stimulating agent hyporesponsiveness. Nephrology. 2007;12:321–30.PubMedCrossRef
136.
137.
Zurück zum Zitat Chow A, Gervasi D, Guo G, Signore P, del Balzo U, Walkinshaw G. Roxadustat enhances degradation of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) in a HIF and INSIG dependent manner. Keystone Symposia; January 19–23 2020; Keystone, CO. Chow A, Gervasi D, Guo G, Signore P, del Balzo U, Walkinshaw G. Roxadustat enhances degradation of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) in a HIF and INSIG dependent manner. Keystone Symposia; January 19–23 2020; Keystone, CO.
139.
Zurück zum Zitat Culleton BF, Walsh M, Klarenbach SW, et al. Effect of frequent nocturnal hemodialysis vs conventional hemodialysis on left ventricular mass and quality of life: a randomized controlled trial. JAMA. 2007;298:1291–9.PubMedCrossRef Culleton BF, Walsh M, Klarenbach SW, et al. Effect of frequent nocturnal hemodialysis vs conventional hemodialysis on left ventricular mass and quality of life: a randomized controlled trial. JAMA. 2007;298:1291–9.PubMedCrossRef
140.
Zurück zum Zitat Chan CT, Floras JS, Miller JA, Richardson RM, Pierratos A. Regression of left ventricular hypertrophy after conversion to nocturnal hemodialysis. Kidney Int. 2002;61:2235–9.PubMedCrossRef Chan CT, Floras JS, Miller JA, Richardson RM, Pierratos A. Regression of left ventricular hypertrophy after conversion to nocturnal hemodialysis. Kidney Int. 2002;61:2235–9.PubMedCrossRef
141.
Zurück zum Zitat 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. 2020;24:115–25.PubMedCrossRef 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. 2020;24:115–25.PubMedCrossRef
142.
Zurück zum Zitat Caltabiano S, Cizman B, Burns O, et al. Effect of renal function and dialysis modality on daprodustat and predominant metabolite exposure. Clin Kidney J. 2019;12:693–701.PubMedCrossRefPubMedCentral Caltabiano S, Cizman B, Burns O, et al. Effect of renal function and dialysis modality on daprodustat and predominant metabolite exposure. Clin Kidney J. 2019;12:693–701.PubMedCrossRefPubMedCentral
Metadaten
Titel
Burden of Anemia in Chronic Kidney Disease: Beyond Erythropoietin
verfasst von
Ramy M. Hanna
Elani Streja
Kamyar Kalantar-Zadeh
Publikationsdatum
29.10.2020
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 1/2021
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-020-01524-6

Weitere Artikel der Ausgabe 1/2021

Advances in Therapy 1/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.