Roxadustat: First Global Approval
Sohita Dhillon1
Abstract
Roxadustat (Ai Rui Zhuo® in China) is an orally administered, small molecule hypoxia-inducible factor (HIF) prolyl hydroxylase inhibitor that is being developed by FibroGen, in collaboration with Astellas and AstraZeneca, for the treatment of anaemia in patients with dialysis-dependent chronic kidney disease (CKD), non-dialysis-dependent CKD and in patients with myelodysplastic syndromes. The drug reversibly binds to and inhibits HIF-prolyl hydroxylase enzymes that are responsible for the degradation of transcription factors in the HIF family under normal oxygen conditions. Inhibition of these enzymes reduces HIF breakdown and promotes HIF activity, leading to an increase in endogenous erythropoietin production, thereby enhancing erythropoiesis. It also reduces the expression of the peptide hormone hepcidin, improves iron availability and increases haemoglobin levels. HIF regulates the expression of genes in response to reduced oxygen levels, including genes required for erythropoiesis and iron metabolism. Roxadustat is approved in China and is under regulatory review in Japan for the treatment of anaemia in patients with dialysis-dependent CKD. Studies are underway to investigate long-term cardiovascular outcomes with roxadustat versus placebo (for non-dialysis-dependent CKD) or standard of care (for dialysis-dependent CKD). This article summarizes the milestones in the development of roxadustat leading to this first approval.
1 Introduction
Anaemia is a common complication of chronic kidney dis-ease (CKD) that increases in prevalence with the progression of disease [1, 2]. The development of anaemia is largely attributed to the decreased production of renal erythropoietin (EPO) and other factors including iron deficiency (which may be partly related to increased levels of hepcidin), blood loss, reduced erythrocyte survival duration and inflammation [2–4]. Hypoxia-inducible factor (HIF) is a heterodimeric transcription factor responsible for the hypoxic induction of EPO and other oxygen-sensitive genes. HIF-prolyl hydroxylase (HIF-PHD) enzymes regulate the stability of the HIF-α subunit of the HIF transcription factor by post-translational hydroxylation of HIF in an oxygen-dependent manner, thus maintaining a balance between oxygen availability and HIF
This profile has been extracted and modified from the AdisInsight database. AdisInsight tracks drug development worldwide through the entire development process, from discovery, through pre-clinical and clinical studies to market launch and beyond.
Sohita Dhillon [email protected]
1 Springer, Private Bag 65901, Mairangi Bay, 0754 Auckland, New Zealand
activity. The central role of HIF-PHD enzymes as gatekeepers of coordinated transcriptional adaptation to hypoxia and oxidative stress make them attractive therapeutic targets for the treatment of anaemia [2, 5].
Several novel orally active small molecules have been developed that transiently inhibit HIF-PHD enzymes, leading to the accumulation of functional HIF, an increase in endogenous EPO production (resulting in enhanced erythropoiesis) and indirect suppression of hepcidin [2, 5]. Roxadustat (Ai Rui Zhuo® in China) is a first-in-class potent HIF-PHD inhibitor being developed by FibroGen, in collaboration with Astellas and AstraZeneca, for the treatment of anaemia in patients with dialysis-dependent CKD, nondialysis-dependent CKD and myelodysplastic syndromes. Roxadustat received its first global approval in China on 17 December 2018 for the treatment of anaemia caused by CKD in patients who are dialysis-dependent [6].
1.1 Company Agreements
In September 2004, FibroGen entered into an agreement with Yamanouchi to license FG 2216 and other related compounds, including roxadustat. Yamanouchi acquired exclusive rights to develop and market the compounds for the treatment of anaemia in Japan, while FibroGen retained
CFDA accepts NDA for DD-CDK and NDD-CDK anaemia (Oct)
CFDA grants priority review status (Oct)
Phase 1 clinical trials initiated (Nov 2005) Phase 2 clinical trials initated (Dec 2006)
Intention to submit MAA to EMA announced (Feb)
Intention to submit NDA to US FDA announced (Jun)
Approved in China (Dec)
2012 2013 2014 2015 2016 2017 2018
NCT02273726 (SIERRAS) NCT02652806
NCT02952092 NCT02780726
NCT02174731 (ROCKIES) NCT02278341 (PYRENEES)
Key milestones in the development of roxadustat for the treatment of anaemia caused by chronic kidney disease in patients who are dialysis dependent, with the focus on phase 3 studies. CFDA China Food and Drug Administration, DD-CKD dialysis-dependent chronic kidney disease, NDA new drug application, NDD-CKD non-dialysis-dependent chronic kidney disease
the rights for the rest of the world. Yamanouchi merged with Fujisawa in April 2005 to form Astellas Pharma [7]. FibroGen has since licensed its oral HIF-PHD inhibitors, including FG 2216 and roxadustat to Astellas Pharma for additional markets including, Europe, the Commonwealth of Independent States, the Middle East and South Africa. FibroGen retained rights in the rest of the world, except Japan (where Astellas already has rights). The agreement was for both the companies to share leadership of the clini-cal development programme in Europe and North America. Under the terms of the agreement, Astellas would pay a licensing fee of $300 million and development milestones totalling $465 million and will share costs of a transatlantic development programme and patent support [8]. Astellas made a payment of $US40 million to FibroGen based on the advancement of roxadustat into phase 2b testing for anaemia associated with CKD in the fourth quarter of 2010 [9].
In July 2013, AstraZeneca and FibroGen entered into a collaboration agreement for the development and com-mercialisation of roxadustat for the treatment of anaemia associated with CKD and end-stage renal disease (ESRD), and potentially other anaemia indications. The companies collaborated on the development in the USA, China and all major markets except those that are covered by the agreement with Astellas (i.e. Japan, Europe, the Commonwealth of Independent States, the Middle East and South Africa)
[10, 11]. Agreements between FibroGen and AstraZeneca include milestone and tiered royalty payments [12]. The agreement is that AstraZeneca is responsible for the US commercialisation of roxadustat, and FibroGen will undertake specified promotional activities in the ESRD segment in this market. After marketing approval, FibroGen China will be responsible for management of manufacturing and medical affairs, while AstraZeneca will oversee promotional activities and commercial distribution [13]. Additional development milestones will be payable for any subsequent indications which the companies choose to pursue. In July 2016, FibroGen received a scheduled license payment of
$US62 million under its collaboration agreement with AstraZeneca [11]. In June 2015, FibroGen received $US120 million non-contingent license payment from AstraZeneca under its collaboration agreements [10, 12, 13].
1.2 Patent Information
As of February 2018, FibroGen holds multiple US patents and some patents in China for roxadustat, its composition of matter, pharmaceutical compositions containing roxadustat and for methods for treating anaemia using roxadustat or its analogues. These patents are valid through 2024 or 2025. The patents for crystalline form of roxadustat in the US and China are set to expire in 2033 [14].
O
OH
N
H O
O
Chemical structure of roxadustat
2 Scientific Summary
2.1 Pharmacodynamics
Roxadustat reversibly binds to and potently inhibits HIF-PHD enzymes, reducing HIF-α breakdown and promoting HIF transcriptional activity [15]. Activation of the HIF pathway in this manner results in the induction of target genes involved in erythropoiesis, such as those for EPO, EPO receptor, proteins promoting iron absorption, iron transport and haem synthesis [15, 16]. Roxadustat dose-dependently increased haemoglobin (Hb) levels (significant difference from placebo at higher doses), significantly reduced hepcidin levels and transiently increased endogenous EPO levels within or near physiological range in patients with anaemia of CKD who were not dialysis dependent [15]. Roxadustat reduced the dysregulation of iron metabolism associated with CKD by increasing serum transferrin, intestinal iron absorption and the release of stored iron in a dose-dependent manner in patients with anaemia associated with dialysis-dependent or dialysis-independent CKD [15, 17]. Cholesterol levels were also significantly reduced from baseline with roxadustat [17, 18], regardless of the use of statins or other lipid-lowering agents [18].
2.2 Pharmacokinetics
The pharmacokinetics of roxadustat have been evaluated in 27 Caucasian and 30 Japanese subjects in two open-label, single-arm, phase 1 dose-escalation studies (FGCL-SM4592-016 and 1517-CL-0201 A/C, respectively). In healthy Caucasian subjects receiving single oral doses of roxadustat 0.3, 1, 2, 3 or 4.0 mg/kg, the mean peak drug
concentrations (Cmax) were 1, 5, 11, 17 and 26 µg/mL, respectively, the mean area under the concentration-time curve (AUC) values were 8, 27, 80, 126 and 219 h ∙ µg/mL
and the mean half-life values were 12, 12, 14, 10 and 12 h (data on file). Overall, the pharmacokinetics of roxadustat in healthy Japanese subjects receiving single-dose roxadustat 1, 2, 3 or 4 mg/kg were generally similar to those in Caucasian subjects [19]. No drug accumulation was observed after repeated thrice weekly dosing of roxadustat [19].
In 75 Chinese healthy volunteers receiving single or multiple thrice-weekly doses of oral roxadustat in two ran-domized, double-blind, placebo-controlled studies, a dose-proportional increase in plasma exposure (AUCτ and Cmax) to roxadustat was observed over a dose range of 40–200 mg [20]. Following single or multiple oral doses of roxadustat 40, 100, 160 or 200 mg, roxadustat mean Cmax of 4, 8, 14 and 15 µg/mL, respectively, was reached within a median time of 2–3 h and the mean elimination half-life was 7–9 h across the treatment groups. No significant drug accumulation was seen after 2 weeks’ treatment with thrice-weekly roxadustat [20].
No significant effect of food was seen on the pharma-cokinetics of roxadustat in an open-label, randomized, phase 1 crossover study (NCT02805374) in 16 healthy adult Japanese subjects [21]. Moderate hepatic impairment is not expected to affect the pharmacokinetics of roxadustat to a clinically significant extent in an open-label phase 1 study in patients with moderate hepatic impairment or normal hepatic function (n = 8/group) [22]. Coadministra-tion of roxadustat with spherical carbon adsorbent (used to reduce the concentration of uremic toxins in systemic circulation; NCT02693613) [21], omeprazole (1517-CL-0527; EudraCT 2014-000666-22) [23] or lanthanum carbonate hydrate (used for the treatment of hyperphosphataemia in patients with end-stage CKD; NCT02952040) [24] did not have a clinically significant effect on the pharmacokinetics of roxadustat in open-label phase 1 studies in healthy subjects. Another open-label phase 1 study in healthy volunteers (NCT02252731) showed that the roxadustat had no clinically significant effect on the pharmacokinetics and limited effect on the pharmacodynamics of warfarin when the two drugs were coadministered [25].
2.3 Therapeutic Trials
2.3.1 In Dialysis‑Dependent Patients with CKD
2.3.1.1 Phase 3 Studies Several phase 3 studies assess-ing the efficacy of roxadustat in patients with anaemia and dialysis dependent CKD have recently been completed and initial data are available; full publication of these data are awaited. In the randomized, open-label, phase 3 SIERRAS trial (NCT02273726; n = 741), roxadustat was noninferior to epoetin alfa for the treatment of anaemia (i.e. maintaining Hb levels) in patients with CKD who were dialysis dependent and were receiving stable doses of erythropoiesis stimulating agent (ESA) [26]. The mean treatment duration was
1.9 years and mean baseline Hb levels were 10.3 g/dL in both treatment groups. For the US primary endpoint, the mean change in Hb from baseline to the average over weeks 28–52 was 0.39 g/dL for roxadustat versus − 0.09 g/dL for epoetin alfa [least squares (LS) mean treatment difference 0.48 g/
Alternative names ASP 1517; AZD 9941; FG-4592
Class Amide; antianaemic; carboxylic acid; isoquinoline; small molecule
Mechanism of action Binds to and potently inhibits HIF-PHD, reducing HIF breakdown and promoting its activity Route of administration Oral
Pharmacodynamics Dose-dependently increased Hb levels, reduced hepcidin and cholesterol levels and transiently increased endogenous EPO levels within or near physiologic range
Pharmacokinetics Exposure increased dose-dependently (approximately proportional to dose) across 1–4 mg/kg dose range; half-life was 8–12 h
Most frequent adverse events Nasopharyngitis, back pain, diarrhoea, vomiting, decreased appetite, muscle spasms, hypertension, decreased transferrin saturation, headache, fatigue, hyperkalaemia
ATC codes
WHO ATC code B03X-A05
EphMRA ATC code B3
Chemical name (((4-Hydroxy-1-methyl-7-phenoxyisoquinolin-3- yl)carbonyl)amino)acetic acid
EPO erythropoietin, Hb haemoglobin, HIF hypoxia-inducible factor, HIF-PHD hypoxia-inducible factor prolyl hydroxylase enzyme
dL; 95% CI 0.37–0.59] and for the EU primary endpoint, the mean change in Hb from baseline to the average over weeks 28–36 was 0.54 versus − 0.02 g/dL (LS mean treatment difference 0.53 g/dL; 95% CI 0.39–0.67). The lower margins of the 95% CIs of the LS mean treatment differences were above the non-inferiority margin of − 0.75 g/dL for both the US and EU primary endpoints, indicating the noninferiority of roxadustat and epoetin alfa. Subsequent superiority testing showed that roxadustat significantly increased Hb levels relative to epoetin alfa, as assessed by the US and EU primary efficacy endpoints (both p < 0.0001). In addition, for the prespecified secondary efficacy analysis of the time to first blood transfusion during treatment, roxadustat significantly (p = 0.0337) reduced the risk of blood transfusion by 33% compared with epoetin alfa [hazard ratio (HR) 0.67] [26].
Roxadustat was no less effective than epoetin alfa in treating anaemia in a phase 3 study in Chinese patients with CKD on haemodialysis or peritoneal dialysis with previous ESA therapy for > 6 weeks (NCT02652806; n = 304) [27]. At baseline, the average Hb was 10.4 g/dL. Following treatment with roxadustat or epoetin alfa thrice weekly for 26 weeks (titrated to maintain/achieve Hb 10–12 g/dL), the average Hb change from baseline to weeks 23–27 with roxadustat was noninferior to that with epoetin alfa (primary endpoint) in the per protocol population (0.8 vs. 0.5 g/dL) and in the full analysis set (no quantitative data available). Superiority of roxadustat over epoetin alfa was demonstrated in the per protocol set (p = 0.037), but not in the full analysis set. Roxadustat also significantly increased transferrin levels, maintained serum iron and attenuated decreases in transferrin saturation (TSAT) relative to epoetin alfa (all p < 0.01). In addition, hepcidin levels were significantly reduced from baseline with roxadustat (by 30.2 ng/mL; p = 0.028
vs. baseline) compared with 2.3 ng/mL with epoetin alfa (p = non significant) [27].
Roxadustat maintained Hb levels and was no less effective than darbepoetin alfa in a randomized, double-blind, active comparator-controlled, phase 3 conversion study in Japanese CKD patients with anaemia on haemodialysis for ≥ 12 weeks and treated with ESA (NCT02952092; n = 303) [28]. Patients received roxadustat (70 or 100 mg) thrice weekly or darbepoetin alfa (10–60 µg) once weekly, with the dosages adjusted to maintain Hb levels of 10–12 g/ dL. The average Hb at weeks 18–24 was 11 g/dL (95% CI 10.88–11.10), confirming the efficacy of roxadustat in maintaining Hb levels as the 95% CIs of the average were between the predefined margin of 10–12 g/dL (primary endpoint). Roxadustat was noninferior to darbepoetin alfa as the lower limit of the 95% CI of the difference between roxa-dustat and darbepoetin alfa was more than the noninferiority criterion of − 0.75 g/dL [28].
Roxadustat achieved and maintained target Hb levels (two thresholds of 10 and 10.5 g/dL; and Hb increase ≥ 1 g/dL) in an open-label, multicentre, phase 3 study in Japanese CKD patients with anaemia on peritoneal dialysis (NCT02780726; n = 56) [29]. Patients not previously treated with ESA (n = 13) were randomized to 50 or 70 mg roxadustat, and those previously treated with ESA (n = 43) were switched to 70 or 100 mg roxadustat depending on previous ESA dose; dosages were adjusted to maintain Hb levels of 10–12 g/dL. The Hb maintenance rate with roxadustat in ESA untreated patients was 92% and in ESA treated patients was 74%. The maintenance rate in patients with at least one Hb target value at weeks 18–24 in ESA untreated patients was 92% and in ESA treated patients was 87% [29].
Roxadustat significantly improved Hb levels from baseline averaged over weeks 28–52 relative to epoetin alfa in
the randomized, open-label, multicentre, active compar-ator-controlled, phase 3 ROCKIES study in patients with anaemia in CKD who were on haemodialysis or peritoneal dialysis (NCT02174731; n = 2133) [30]. The study included patients who had been treated with an erythropoietin ana-logue or have an indication for treatment with an erythro-poietin analogue.
2.3.1.2 Phase 2 Studies Roxadustat was at least as effective as epoetin alfa in maintaining Hb levels in two randomized, open-label phase 2 studies in patients with anaemia and ESRD who were dependent on dialysis [17, 31]. In one study (NCT01596855; n = 87), patients who had received stable doses of intravenous or subcutaneous epoetin alfa for 7 weeks prior to randomization were randomized to receive roxadustat thrice weekly at low (1.1–1.8 mg/kg), medium (1.5–2.3 mg/kg) or high (1.7–2.3 mg/kg) starting doses or to the continuation of epoetin alfa [17]. Patients had to have a mean Hb level of 9–12 g/dL in three screening tests prior to randomization. The primary endpoint was the proportion of patients with successful dose conversion, i.e. an Hb level maintained at no less than 0.5 g/dL below mean baseline value in the last 2 weeks of the 6-week dosing period in the efficacy evaluable population. Following 6 weeks of therapy, 59, 89 and 100% of patients receiving roxadustat low, medium and high doses, respectively, met the primary endpoint compared with 50% of patients who continued epoetin alfa therapy (p = 0.008 for mid-dose and p = 0.0003 for high-dose roxadustat vs. epoetin alfa) [17].
The other study (NCT01147666) had two parts and
included patients who were previously maintained with intravenous epoetin alfa and intravenous iron for 4 weeks prior to randomization and had a mean Hb level of 9–13.5 g/ dL for 8 weeks [31]. Part 1 was a 6-week dose-ranging study in 54 patients receiving oral roxadustat doses thrice weekly (1–2 mg/kg fixed doses) versus continuation of intravenous epoetin alfa. Hb level responder rates were significantly (p = 0.03) higher in patients receiving roxadustat 1.5–2 mg/kg (pooled) than in those receiving epoetin alfa (79 vs. 33%; primary endpoint). Response rate was defined as the proportion of participants whose Hb levels did not decrease by > 0.5 g/dL from baseline. In addition, the hepcidin level was significantly (p < 0.05) reduced with roxadustat 2 mg/kg than with epoetin alfa [31].
In part 2 (19 weeks treatment), 90 patients in six cohorts received roxadustat thrice weekly at various starting doses and adjustment rules (1.0–2.0 mg/kg or tiered weight-based; n = 67) or continued epoetin alfa therapy (n = 23) [31]. In the efficacy-evaluable population, 51% (31/61) of patients receiving roxadustat achieved average Hb levels of ≥ 11 g/dL over the last 4 weeks of the 19-week treatment period compared with 36% (8/22) of patients receiving epoetin alfa
(primary endpoint). Roxadustat and epoetin alfa recipients did not significantly differ in the change from baseline in Hb levels at any timepoint over 19 weeks of therapy (LS mean change − 0.5 g/dL in the pooled roxadustat and epoetin alfa groups; LS mean treatment difference − 0.03 g/dL; 95% CI
– 0.39 to 0.33) [31].
2.3.2 In Newly‑Initiated Dialysis Patients
In the randomized, open-label, active comparator-controlled, phase 3 HIMALAYAS trial (NCT02052310; n = 1043), roxadustat was noninferior to epoetin alfa for the treatment of anaemia (as assessed by the US and EU primary endpoints) in patients who had newly initiated dialysis treatment for ESRD [26]. The mean treatment duration was 1.8 years and the mean baseline Hb level was approximately 8.4 g/dL. For the US primary endpoint, the mean change in Hb from baseline to the average over weeks 28–52 was 2.57 g/dL for roxadustat versus 2.36 g/dL for epoetin alfa (LS mean treatment difference 0.18 g/dL; 95% CI 0.08–0.29). The lower margin of the 95% CI of the LS mean treatment difference was above the non-inferiority margin of − 0.75 g/dL, indicating the noninferiority of roxadustat and epoetin alfa. Subsequent superiority testing showed that roxadustat significantly increased Hb levels relative to epoetin alfa as assessed by the US primary endpoint (p = 0.0005). Noninferiority between roxadustat and epoetin alfa was also demonstrated for the EU primary endpoint of the propor-tion of patients achieving an Hb response at 24 weeks (88 vs. 85%); the lower margin of the 95% CI (− 0.9 to 7.6%) for the between-group difference was above the non-inferiority margin of − 15%. Hb response was defined as an Hb level of
≥ 11 g/dL and an Hb increase of ≥ 1 g/dL [26].
Roxadustat corrected anaemia in CKD patients with newly initiated haemodialysis or peritoneal dialysis in a phase 2b proof-of-concept study (NCT01414075; n = 60) [32]. Patients (baseline Hb 8.3 g/dL) received 12 weeks’ treatment with roxadustat thrice weekly according to body weight (40–60, > 60–90 and > 90–140 kg), with the doses titrated based on Hb response (60–140, 100–200 and 140–300 mg for the respective groups). Roxadustat increased Hb levels by ≥ 2 g/dL within 7 weeks of therapy, regardless of baseline iron repletion status, C-reactive protein level or iron supplementation regimen (oral or intravenous). In the efficacy evaluable population (n = 55), the mean maximal change from baseline in Hb over 12 weeks was 3 g/dL (primary endpoint). TSAT levels, reticulocyte Hb and serum iron levels did not significantly differ from baseline levels in patients receiving oral or intravenous iron supplementation (n = 32), but decreased significantly in patients receiving no iron (all p < 0.05 vs. baseline). [32].
Drug(s) Indication Phase Status Location(s) Identifier Sponsor
Roxadustat, epoetin alfa CKD anaemia in stable 3 Completed USA, Puerto Rico NCT02273726; FGCL- FibroGen
dialysis pts Roxadustat, epoetin alfa CKD anaemia in stable
dialysis pts
4592-064; SIERRAS
3 Completed China NCT02652806; FGCL-4592-806
FibroGen
Roxadustat, darbepoetin alfa
CKD anaemia in ESA-naïve HD pts
3 Completed Japan NCT02952092; 1517-CL-0307
Astellas Pharma
Roxadustat CKD anaemia in PD pts 3 Completed Japan NCT02780726; 1517-CL-
0302
Astellas Pharma
Roxadustat, epoetin alfa CKD anaemia in pts on HD
or PD
3 Completed Worldwide NCT02174731; D5740C00002; ROCKIES
AstraZeneca
Roxadustat, epoetin alfa, darbepoetin alfa
CKD anaemia in ESRD pts on stable dialysis
NCT02278341; 1517-CL-0613; 2013-001497-16; PYRENEES
Astellas Pharma Europe B.V.
Roxadustat, epoetin alfa CKD anaemia in pts with
newly initiated dialysis for ESRD
Roxadustat, placebo CKD anaemia in pts not on
dialysis
Roxadustat, placebo CKD anaemia in pts not on
dialysis
NCT02052310; FGCL-4592-063; 2013-002753-
30: HIMALAYAS
NCT01750190; FGCL-4592-060; ANDES
NCT02174627; D5740C00001; OLYMPUS
FibroGen
FibroGen AstraZeneca
Roxadustat, darbepoetin alfa
Roxadustat, darbepoetin alfa
CKD anaemia in pts not on dialysis
CKD anaemia in pts not on dialysis
NCT02021318; 1517-CL-0610; 2013-000951-42; DOLOMITES
NCT02988973; 1517-CL-0310
Astellas Pharma Europe B.V.
Astellas Pharma
Roxadustat, placebo CKD anaemia in pts not on
dialysis
Roxadustat, placebo CKD anaemia in pts not on
dialysis
NCT02652819; FGCL-4592-808
NCT01887600; 1517-CL-0608; 2012-005180-27; ALPS
FibroGen
Astellas Pharma Europe B.V.
Roxadustat, placebo Primary MDS 3 Recruiting Worldwide NCT03263091; FGCL-
4592-082
Roxadustat, placebo Primary MDS 2/3 Recruiting China NCT03303066; FGCL-
4592-813
FibroGen FibroGen
Roxadustat CKD anaemia in dialysis and nondialysis pts
2/3 Unknown USA, Puerto Rico NCT01630889; FGCL-
4592-059
FibroGen
Roxadustat CKD anaemia incident to HD or PD
2b Completed USA, Hong Kong,
Russia, Singapore
NCT01414075; FGCL-4592-053
FibroGen
Roxadustat, epoetin alfa CKD anaemia in HD pts 2
with ESRD Completed China NCT01596855; FGCL- FibroGen 4592-048
Roxadustat, epoetin alfa CKD anaemia in HD pts 2
with ESRD Completed USA NCT01147666; FGCL- FibroGen 4592-040
Roxadustat, darbepoetin CKD anaemia in pts on 2
alfa dialysis Completed Japan NCT01888445; 1517-CL- Astellas Pharma 0304
Roxadustat, placebo CKD anaemia in pts not on 2
dialysis Completed China NCT01599507; FGCL- FibroGen 4592-047
Roxadustat, placebo CKD anaemia in pts not on 2
dialysis Completed Japan NCT01964196; 1517-CL- Astellas Pharma 0303
Roxadustat CKD anaemia in pts not on 2
dialysis Completed USA, Puerto Rico NCT01244763; FGCL- FibroGen 4592-041
Roxadustat CKD anaemia in pts not on 2 Completed USA NCT00761657; FibroGen
dialysis FGCLSM4592-017
CKD chronic kidney disease, ESA erythropoiesis stimulating agent, ESRD end-stage renal disease, ID incident dialysis, HD haemodialysis, MDS
myelodysplastic syndrome, PD peritoneal dialysis, pts patients
2.3.3 In Non‑Dialysis Patients
Several phase 3 studies assessing the efficacy of roxadustat in patients with anaemia and non-dialysis-dependent CKD have been completed and initial data are available; fully published data are awaited. In the randomized, double-blind, placebo-controlled, phase 3 ANDES trial (NCT01750190; n = 922), roxadustat was effective for the treatment of anaemia (as assessed by the US and EU primary endpoints) in patients with later-stage CKD (stages 3, 4 or 5) who were not dialysis-dependent [26]. The average treatment duration was 1.7 years and the average baseline Hb level was 9.1 g/dL in both, roxadustat and placebo, groups. For the US primary endpoint, the mean change in Hb from baseline to the average over weeks 28–52 was significantly higher with roxadustat than with placebo (2.0 vs. 0.16 g/dL; p < 0.0001). For the EU primary endpoint, a higher proportion of roxadustat than placebo recipients achieved an Hb response (86 vs. 7%; p = 0.0007), defined as an Hb level of ≥ 11 g/dL and an Hb increase of ≥ 1 g/dL. In a prespecified secondary analysis, roxadustat significantly reduced the risk of rescue therapy by 81% relative to placebo (HR 0.19; p < 0.0001), where rescue therapy was defined as administration of ESA or intravenous iron in the first 52 weeks of treatment. In terms of the time to first blood transfusion in the first 52 weeks of treatment, roxadustat reduced the risk of blood transfusion by 74% (HR 0.26; p < 0.0001) [26].
The randomized, double-blind, placebo-controlled phase
3 ALPS study (NCT01887600) in patients with anaemia of CKD who were not receiving dialysis demonstrated the superiority of roxadustat both in terms of Hb response rate in the first 24 weeks of treatment and Hb change from baseline at weeks 28–52 (coprimary endpoints) [33].
In the randomized, double-blind, placebo-controlled, phase 3 OLYMPUS study (NCT02174627; n = 2781), sig-nificant and clinically relevant improvement from baseline in Hb levels averaged over weeks 28–52 was seen with roxadustat relative to placebo in patients with anaemia and CKD stages 3, 4 or 5 whose disease progression was moderate to severe and who were not dialysis dependent [30].
Roxadustat was effective in correcting and maintaining Hb levels in a randomized, double-blind, placebo-controlled phase 3 study in Chinese patients with anaemia (baseline Hb 7–10 g/mL) and CKD who were not dialysis dependent (NCT02652819; n = 152 safety population) [34]. Following 8 weeks treatment with roxadustat thrice weekly or pla-cebo, the mean change from baseline in Hb levels was sig-nificantly greater with roxadustat than with placebo (1.9 vs.
– 0.39 g/dL; p < 0.000000000000001). At week 9, roxadustat recipients had significantly greater reduction from baseline in hepcidin levels (mean change − 56 vs. − 15 ng/mL; p < 0.00001) and lipid fractions (total and low density lipoprotein cholesterol; p < 0.00001) [34].
In an earlier randomized, open-label, multicentre, phase 2b study (NCT01244763), roxadustat administered at various starting doses and frequencies for 16 or 24 weeks achieved anaemia correction and reduced serum hepcidin levels in 143 evaluable patients with anaemia (baseline Hb ≤ 10.5 g/dL) and CKD not dependent on dialysis [18]. After 16 weeks’ roxadustat treatment, 92% of patients overall had achieved an Hb response, defined as the proportion of patients with Hb increase of ≥ 1.0 g/dL from baseline and Hb of ≥ 11.0 g/dL by the end of week 16 (primary endpoint). Treatment with roxadustat significantly reduced hepcidin levels by 17% (p = 0.004), increased Hb levels by a mean of 1.83 g/dL (p < 0.001) and maintained reticulocyte Hb content over the first 16 weeks of treatment. Mean total cholesterol was also significantly (p < 0.001) reduced by 26 mg/dL with roxadustat after 8 weeks’ treatment, regardless of the use of statins or other lipid-lowering agents [18]. In a randomized, double-blind phase 2 study in patients with anaemia and CKD who were not dialysis dependent (NCT01599507; n = 91), significantly more patients receiving low-dose (1.1–1.75 mg/kg) or high-dose (1.5–2.25 mg/kg) roxadustat thrice weekly for 8 weeks than those receiving placebo had an Hb increase of ≥ 1 g/dL from baseline (80 and 87% vs. 23%; both p < 0.0001) [17]. A randomized, single-blind, placebo-controlled phase 2a study (NCT00761657; n = 116) in a similar patient population showed that roxadustat (0.7, 1.0, 1.5 or 2 mg/kg) thrice weekly increased Hb levels in a dose-dependent manner (0.8–2.2 mg/dL) during the 6-week study period (4 weeks treatment), with a significant difference between the 1.5 and
2.0 mg/kg treatment groups versus the placebo group (mean
change + 1.2 and + 1.8 vs. − 0.1 g/dL; both p < 0.01) [15].
2.4 Adverse Events
Data for the tolerability of roxadustat thrice weekly in phase 3 clinical trials of anaemia of CKD are limited as of January 2019. Its tolerability profile in preliminary safety analyses of phase 3 clinical trials in patients with anaemia and dialysis-dependent CKD (SIERRAS), non-dialysis CKD (ANDES) and incident dialysis CKD (HIMALAYS) was consistent with that expected in study populations with similar background diseases (no quantitative data currently available) [26]. Roxadustat was generally well tolerated in phase 3 studies in Chinese [27, 34] and Japanese [28, 29] patients with dialysis-dependent [27–29] or non-dialysis
[34] CKD. In two phase 3 studies in Japanese patients with anaemia and dialysis-dependent CKD, the most common treatment-emergent adverse events (AEs) with roxadustat included nasopharyngitis, back pain, diarrhoea, vomiting [28, 29]; its tolerability profile was generally similar to that of darbepoetin alfa [28]. Pooled safety data, including major adverse cardiovascular (CV) events, in patients with
dialysis-dependent or non-dialysis CKD from studies in the global phase 3 program are awaited with interest [26].
In phase 2 studies in dialysis-dependent patients, the tolerability profile of roxadustat thrice weekly was con-sistent with the background disease and generally similar to that of epoetin alfa [17, 31] In a dose-ranging study in Chinese ESRD patients who were dependent on dialy-sis and previously had Hb levels maintained with epoetin alfa (NCT01147666), treatment-emergent AEs occurred in 64% (69/108) of roxadustat compared with 61% (22/36) of epoetin alfa recipients in the safety population follow-ing ≤ 19 weeks’ treatment [31]. Serious AEs (SAEs) were reported in 24% of patients in the roxadustat group and 17% of patients in the placebo group. A post hoc, exploratory composite CV safety event (comprising death, myocardial infarction, stroke, heart failure requiring hospitalization, unstable angina requiring hospitalization or thromboem-bolism in patients receiving 19 weeks’ therapy) occurred in 12% of roxadustat compared with 17% of epoetin alfa. Three deaths were reported in the roxadustat group, none of which were considered treatment related [31]. Generally similar results were seen in another phase 2 study conducted in China in a similar patient population (NCT01596855) [17]. Treatment-emergent AEs were reported in 43% (32/74) of roxadustat and 18% (4/22) of epoetin alfa recipients, with the most common (incidence ≥ 5%) events with roxadustat being decreased appetite (7 vs. 5% with epoetin alfa) and muscle spasms (5 vs. 14%). No SAEs were reported in this study [17].
Roxadustat was generally well tolerated in a phase
2b study in patients with newly initiated dialysis (NCT01414075) [32]. Treatment-emergent AEs occurred in 50% (30/60) of patients receiving roxadustat thrice weekly for 12 weeks, with severe, life-threatening or fatal AEs reported in 10% of patients. The most common (incidence
> 5%) AEs with roxadustat were hypertension necessitating an increase or change of antihypertensive medication (10%; four patients on haemodialysis and two patients on peritoneal dialysis) and a decrease in TSAT (7%). Treat-ment emergent SAEs occurred in 12% of patients and there were two deaths, both of which were considered unrelated to roxadustat therapy [32].
During 12 weeks’ follow-up (4 weeks treatment) in a phase 2a study in patients with anaemia and CKD who were not dialysis dependent (NCT00761657), 59% (52/88) of roxadustat (0.7, 1.0, 1.5 or 2 mg/kg) thrice weekly recipients compared with 46% (13/28) of placebo recipients had treatment-emergent AEs [15]. The most common AEs with roxadustat were expected in patients with CKD and did not differ from those occurring in placebo recipients to a clinically relevant extent. These included diarrhoea (9 vs. 7% with placebo), headache (7 vs. 4%), back pain (5 vs. 4%), fatigue (5 vs. 0%) and hyperkalaemia (5 vs. 0%). SAEs were reported
in four patients receiving roxadustat (vascular access complications, femoral neck fracture, noncardiac chest pain and dyspnoea) and one patient receiving placebo. There were no reports of treatment-related exacerbation of hypertension, seizures, thromboembolic and CV events with roxadustat. There were no deaths, CV SAEs or episodes of liver toxicity or sustained increases in liver enzymes or serum bilirubin in the study. An increase in Hb of > 13 g/dL on at least one occasion by the end of week 6 was reported in 13% of roxadustat and 4% of placebo recipients, all of which decreased to < 12 g/dL within 4–8 weeks; none of the episodes were associated with hypertension, seizure or CV event [15].
In another phase 2 study in patients with anaemia and CKD who were not dialysis dependent (NCT01599507), treatment-emergent AEs were reported in 59% (36/61) of patients receiving low-dose (1.1–1.75 mg/kg) or high-dose (1.5–2.25 mg/kg) roxadustat thrice weekly compared with 63% (19/30) of patients receiving placebo [17]. The most common (incidence > 5%) treatment-emergent AEs with roxadustat that occurred more frequently than with pla-cebo were decrease in TSAT (13 vs. 3%), hyperkalaemia (10 vs. 7%), nausea (7 vs. 3%), hypertension (7 vs. 0%) and chronic renal failure (7 vs. 0%). Treatment-emergent SAEs occurred in 13% (8/61) of roxadustat and 13% (4/30) of placebo recipients, none of which were considered treatment related [17]. A third phase 2 study in this patient population (NCT01244763) supported the tolerability profile of roxadustat, with no treatment-related SAEs reported [18].
2.5 Ongoing Clinical Trials
Several phase 3 efficacy and safety trials of roxadustat are ongoing, including the randomized, open-label, active-controlled, phase 3 DOLOMITES study (NCT02021318), which is comparing the efficacy and safety of roxadustat with that of darbepoetin alfa for the treatment of anaemia in 616 patients with CKD who are not on dialysis. The primary endpoint is the Hb response to roxadustat without the use of rescue therapy, and the study is expected to be completed by November 2019. Recruitment is underway in a randomized, open-label, active-controlled, phase 3 Japanese study (NCT02988973) comparing the efficacy and safety of roxadustat with that of darbepoetin alfa in non-dialysis-dependent patients with CKD who have switched from recombinant human EPO or darbepoetin alfa to roxadustat. Another cohort in this study is evaluating the efficacy and safety of roxadustat in patients switched from epoetin beta pegol to roxadustat. The study plans to enrol 325 patients and the primary endpoint is the change from baseline in the average Hb over weeks 18–24; the study is expected to be completed by November 2019. In addition, an open-label phase 2/3 extension study (NCT01630889) is evaluating
the long-term efficacy and safety of roxadustat in maintaining Hb levels in dialysis- and non-dialysis-dependent CKD patients who have completed treatment in a FibroGen-spon-sored anaemia study.
Recruitment is underway in a randomized, double-blind placebo-controlled phase 3 study (NCT03263091) assess-ing the efficacy and safety of roxadustat in the treatment of anaemia in patients with lower risk MDS and low red blood cell transfusion burden. The study plans to enrol 184 patients and the primary efficacy endpoint is the achieve-ment of transfusion independence for ≥ 56 days. Recruit-ment is also underway in a phase 2/3 study (NCT03303066) evaluating the efficacy and safety of roxadustat in Chinese patients with lower risk MDS. The study plans to enroll 175 patients and is expected to be completed by 2020; the primary endpoint of the study is the proportion of subjects with an Hb response to roxadustat without need for transfusion.
3 Current Status
Roxadustat received its first global approval in China on 17 Decem-ber 2018 for the treatment of anaemia caused by CKD in patients who are dialysis-dependent [6]. It can be prescribed to patients who use haemodialysis or peritoneal dialysis [6].
Compliance with Ethical Standards
Funding The preparation of this review was not supported by any external funding.
Conflict of interest During the peer review process the manufacturer of the agent under review was offered an opportunity to comment on the article. Changes resulting from any comments received were made by the authors on the basis of scientific completeness and accuracy. Sohita Dhillon is a salaried employee of Adis/Springer, is responsible for the article content and declares no relevant conflicts of interest.
References
1. Becker K, Saad M. A new approach to the management of anemia in CKD patients: a review on roxadustat. Adv Ther. 2017;34(4):848–53.
2. Locatelli F, Fishbane S, Block GA, et al. Targeting hypoxia-induc-ible factors for the treatment of anemia in chronic kidney disease patients. Am J Nephrol. 2017;45(3):187–99.
3. 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(6):815–26.
4. Fishbane S, Spinowitz B. Update on anemia in ESRD and ear-lier stages of CKD: core curriculum 2018. Am J Kidney Dis. 2018;71(3):423–35.
5. Haase VH. HIF-prolyl hydroxylases as therapeutic targets in erythropoiesis and iron metabolism. Hemodial Int. 2017;21(Suppl 1):S110–24.
6. FibroGen. FibroGen announces approval of roxadustat in China for the treatment of anemia in chronic kidney disease patients on dialysis [media release]. 17 Dec 2018. http://investor.fibrogen. com/phoenix.zhtml?c=253783&p=irol-newsArticle&ID=23809 52.
7. FibroGen. FibroGen enters agreement with Yamanouchi to license erythropoietic small molecule in development for the treatment of anemia [media release]. 24 Sep 2004. http://investor.fibrogen. com/phoenix.zhtml?c=253783&p=irol-newsArticle&ID=19843 18.
8. FibroGen. FibroGen licenses oral HIF-PH inhibitors, including FG-2216 and FG-4592, to Astellas for the treatment of anemia in Europe and other regions; FibroGen positioned to develop first oral anemia therapy for North America [media release]. 28 Apr 2006. http://investor.fibrogen.com/phoenix.zhtml?c=25378 3&p=irol-newsArticle&ID=1984254.
9. FibroGen. FibroGen announces initiation of phase 2b studies of FG-4592, an oral HIF prolyl hydroxylase inhibitor, for treatment of anemia in chronic kidney disease [media release]. 17 Mar 2011. http://investor.fibrogen.com/phoenix.zhtml?c=253783&p=irol-newsArticle&ID=1983440.
10. AstraZeneca. AstraZeneca and FibroGen collaborate to develop and commercialize FG-4592, a treatment for anemia in chronic kidney disease and end-stage renal disease [media release]. 31 Jul 2013. https://www.astrazeneca.com/media-centre/press-relea ses/2013/astrazeneca-fibrogen-fg4592-anaemia-treatment-chron ic-kidney-disease-renal-disease-31072013.html#.
11. FibroGen. FibroGen announces receipt of $62 million license payment from AstraZeneca [media release]. 8 Jul 2016. http://inves tor.fibrogen.com/phoenix.zhtml?c=253783&p=irol-newsArticl e&ID=2182906.
12. FibroGen. FibroGen announces receipt of $120 million license payment from AstraZeneca [media release]. 29 Jun 2015. http:// investor.fibrogen.com/phoenix.zhtml?c=253783&p=irol-newsA rticle&ID=2063398.
13. FibroGen. FibroGen’s roxadustat (FG-4592) meets primary endpoints in two phase 3 anemia studies in China [media release]. 30 Jan 2017. http://investor.fibrogen.com/phoenix.zhtml?c=25378 3&p=irol-newsArticle&ID=2240513.
14. FibroGen Inc. SEC Filings: form 10-K. 2018. http://phx. corporate-ir.net/phoen ix.zhtml ?c=253783&p=irol-SECTe xt&TEXT=aHR0cDovL2FwaS50ZW5rd2l6YXJkLmNvbS9 maWxp bmcue G1sP2 lwYWd lPTEy MDg4M DE4Jk RTRVE 9MCZTRVE9MCZTUURFU0M9U0VDVElPTl9FTlRJUkU
mc3Vic2lkPTU3. Accessed 2018.
15. 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 Transplant. 2015;30(10):1665–73.
16. Haase VH. Hypoxic regulation of erythropoiesis and iron metabolism. Am J Physiol Renal Physiol. 2010;299(1):F1–13.
17. 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 Transplant. 2017;32(8):1373–86.
18. 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(6):982–91.
19. Yu KH, Chou J, Klaus S, et al. Comparable doses of FG-4592 have similar PK/PD in healthy Caucasian and Japanese subjects (abstract no. MP204). Nephrol Dial Transplant. 2013;28(Suppl 1):i362.
20. Chen X, Zheng X, Jiang J, et al. FG4592, a novel inhibitor of the prolyl hydroxylase of hypoxia-inducible factor (HIF-PH) elicited
linear-exponential dose-response profile on plasma erythropoietin (EPO) levels (abstract no. P413). Haematologica. 2013;98(Suppl 1):175–6.
21. 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. 2018. https://doi.org/10.1002/cpdd.597.
22. Groenendaal-van de Meent D, Adel MD, Noukens J, 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(9):743–51.
23. Groenendaal-van de Meent D, den Adel M, van Dijk J, et al. Effect of multiple doses of omeprazole on the pharmacokinetics, safety, and tolerability of roxadustat in healthy subjects. Eur J Drug Metab Pharmacokinet. 2018. https://doi.org/10.1007/s13318-018-0480-z.
24. Shibata T, Nomura Y, Takada A, et al. 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(5):633–9.
25. Groenendaal-van de Meent D, den Adel M, Rijnders S, et al. The hypoxia-inducible factor prolyl-hydroxylase inhibitor roxadustat (FG-4592) and warfarin in healthy volunteers: a pharmacokinetic and pharmacodynamic drug-drug interaction study. Clin Ther. 2016;38(4):918–28.
26. FibroGen. FibroGen announces positive topline results from three global phase 3 trials of roxadustat for treatment of anemia in patients with chronic kidney disease: primary efficacy endpoints met in all three studies, non-dialysis, incident dialysis, and stable dialysis studies [media release]. 20 Dec 2018. http://inves tor.fibrogen.com/phoenix.zhtml?c=253783&p=irol-newsArticl e_print&ID=2381297.
27. Chen N, Hao C-M, Liu B-C, et al. A phase 3, randomized, open-label, active-controlled study of efficacy and safety of roxadustat for treatment of anemia in subjects with CKD on dialysis (abstract no. TH-PO1152). J Am Soc Nephrol. 2018;29(Suppl):B5.
28. Akizawa T, Iwasaki M, Yamaguchi Y, et al. Phase 3, randomized, double-blind, active-comparator (darbepoetin alfa) conversion study of oral roxadustat in CKD patients with anemia on hemo-dialysis in Japan (abstract no. TH-PO1151). J Am Soc Nephrol. 2018;29(Suppl):B4–B5.
29. Akizawa T, Otsuka T, Reusch M, et al. Phase 3, multicenter, open-label study of intermittent oral roxadustat in peritoneal dialysis CKD patients with anemia (abstract no. SA-OR075). J Am Soc Nephrol. 2018;29(Suppl):99.
30. AstraZeneca. Phase III OLYMPUS and ROCKIES trials for roxadustat met their primary endpoints in chronic kidney disease patients with anaemia [media release]. 2018. https://www.astra zeneca.com/media-centre/press-releases/2018/phase-iii-olympus-and-rockies-trials-for-roxadustat-met-their-primary-endpoints-in-chronic-kidney-disease-patients-with-anaemia20122018.html.
31. 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(6):912–24.
32. Besarab A, Chernyavskaya E, Motylev I, et al. Roxadustat (FG-4592): correction of anemia in incident dialysis patients. J Am Soc Nephrol. 2016;27(4):1225–33.
33. Astellas Pharma. Astellas announces positive topline results for global phase 3 trial of roxadustat in chronic kidney disease (CKD) patients with anemia not on dialysis [media release]. 20 Sep 2018. https://www.astellas.com/system/files/news/2018-09/18092 0_eg.pdf.
34. Chen N, Hao C-M, Peng X, et al. A phase 3, randomized, double-blind, placebo-controlled study of efficacy and safety of roxadustat (FG-4592) for treatment of anemia in subjects with CKD not on dialysis (abstract no. TH-PO1153). J Am Soc Nephrol. 2018;29(Suppl.):B5.