Roxadustat in the Treatment of Anemia in End Stage Renal Disease (ESRD) Patients on Stable Dialysis (Pyrenees)

January 14, 2021 updated by: Astellas Pharma Europe B.V.

A Phase 3, Randomized, Open-Label, Active-Controlled Study to Evaluate the Efficacy and Safety of Roxadustat in the Maintenance Treatment of Anemia in End Stage Renal Disease Patients on Stable Dialysis

This study was conducted to explore a new therapy for anemia in participants with end stage renal disease (ESRD) on dialysis. Anemia is a reduced number of red blood cells or hemoglobin. Hemoglobin (which contains iron) is important for the transport of oxygen in your blood. The purpose of this study was to evaluate if roxadustat is effective and safe in the maintenance treatment of anemia in ESRD participants on stable dialysis. Roxadustat was compared to epoetin alfa and darbepoetin alfa, commercially available medicines for treatment of anemia.

Study Overview

Detailed Description

This study consisted of three study periods as follows:

  • Screening Period: up to 6 weeks
  • Treatment Period: a minimum of 52 weeks up to a maximum of 104 weeks
  • Follow-up Period: 4 weeks

Study Type

Interventional

Enrollment (Actual)

838

Phase

  • Phase 3

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

      • Aalst, Belgium, 9300
        • Site BE32001
      • Antwerp, Belgium, 2060
        • Site BE32002
      • Antwerpen, Belgium, 2020
        • Site BE32019
      • Baudour, Belgium, 7331
        • Site BE32012
      • Bonheiden, Belgium, 2820
        • Site BE32017
      • Leuven, Belgium, 3000
        • Site BE32003
      • Liege, Belgium, 4000
        • Site BE32013
      • Roeselare, Belgium, 8800
        • Site BE32011
    • Flemish Brabant
      • Brussels, Flemish Brabant, Belgium, 1200
        • Site BE32004
      • Blagoevgrad, Bulgaria, 2700
        • Site BG35925
      • Haskovo, Bulgaria, 6300
        • Site BG35931
      • Pleven, Bulgaria, 5800
        • Site BG35915
      • Plovdiv, Bulgaria, 4000
        • Site BG35909
      • Plovdiv, Bulgaria, 4003
        • Site BG35919
      • Rousse, Bulgaria, 7002
        • Site BG35920
      • Shumen, Bulgaria, 9700
        • Site BG35938
      • Sofia, Bulgaria, 1309
        • Site BG35924
      • Sofia, Bulgaria, 1431
        • Site BG35906
      • Sofia, Bulgaria, 1527
        • Site BG35921
      • Stara Zagora, Bulgaria, 6000
        • Site BG35907
      • Varna, Bulgaria, 9000
        • Site BG35916
      • Varna, Bulgaria, 9010
        • Site BG35918
      • Veliko Tarnovo, Bulgaria, 5000
        • Site BG35903
      • Yambol, Bulgaria, 8600
        • Site BG35937
      • Cakovec, Croatia, 40000
        • Site HR38508
      • Karlovac, Croatia, 47000
        • Site HR38505
      • Osijek, Croatia, 31 000
        • Site HR38507
      • Rijeka, Croatia, 51000
        • Site HR38506
      • Slavonski Brod, Croatia, 35000
        • Site HR38504
      • Zadar, Croatia, 23 000
        • Site HR38501
    • Grad Zagreb
      • Zagreb, Grad Zagreb, Croatia, 10000
        • Site HR38509
      • Liberec, Czechia, 46063
        • Site CZ42008
      • Praha 6, Czechia, 169 00
        • Site CZ42021
      • Rakovnik, Czechia, 26929
        • Site CZ42015
      • Amiens cedex 1, France, 80054
        • Site FR33005
      • La Tronche, France, 38701
        • Site FR33010
      • Saint Ouen, France, 93400
        • Site FR33055
      • Saint Priez En Jarez, France, 42270
        • Site FR33007
      • Valenciennes, France, 59300
        • Site FR33056
      • Tbilisi, Georgia, 0144
        • Site GE99503
      • Tbilisi, Georgia, 0144
        • Site GE99504
      • Tbilisi, Georgia, 159
        • Site GE99508
      • Berlin, Germany, 10117
        • Site DE49067
      • Cloppenburg, Germany, 49661
        • Site DE49073
      • Dresden, Germany, 01307
        • Site DE49008
      • Dusseldorf, Germany, 40210
        • Site DE49054
      • Frankfurt am Main, Germany, 60590
        • Site DE49020
      • Hamburg, Germany, 23397
        • Site DE49065
      • Heilbronn, Germany, 74076
        • Site DE49075
      • Kaiserslautern, Germany, 67655
        • Site DE49001
      • Muenchen, Germany, 81695
        • Site DE49070
      • Solingen, Germany, 42653
        • Site DE49002
      • Villingen-Schwenningen, Germany, 78052
        • Site DE49071
    • Nordrhein-Westfalen
      • Dormagen, Nordrhein-Westfalen, Germany, 41540
        • Site DE49056
      • Baja, Hungary, 6500
        • Site HU36033
      • Esztergom, Hungary, 2500
        • Site HU36036
      • Gyor, Hungary, 9002
        • Site HU36031
      • Kaposvar, Hungary, H 7400
        • Site HU36026
      • Kistarcsa, Hungary, 2143
        • Site HU36027
      • Pecs, Hungary, 7624
        • Site HU36032
      • Pecs, Hungary, 7633
        • Site HU36035
      • Salgotarjan, Hungary, 3100
        • Site HU36034
      • Szeged, Hungary, 6724
        • Site HU36004
      • Szekesfehervar, Hungary, 8000
        • Site HU36046
      • Szombathely, Hungary, H 9700
        • Site HU36006
      • Zalsaegerszeg, Hungary, 8900
        • Site HU36003
      • Brescia, Italy, 25123
        • Site IT39010
      • Lecco, Italy, 23900
        • Site IT39008
      • Milano, Italy, 20162
        • Site IT39006
      • Modena, Italy, 41124
        • Site IT39037
      • Padova, Italy, 35128
        • Site IT39022
      • Pavia, Italy, 27100
        • Site IT39036
      • Roma, Italy, 122
        • Site IT39005
      • Torino, Italy, 10126
        • Site IT39035
      • Trieste, Italy, 34142
        • Site IT39032
    • Frazione Di Galciana
      • Prato, Frazione Di Galciana, Italy, 59100
        • Site IT39028
    • Lombardia
      • Cremona, Lombardia, Italy, 26100
        • Site IT39039
    • Venezia
      • Mestre, Venezia, Italy, 30174
        • Site IT39014
      • Katowice, Poland, 40 027
        • Site PL48002
      • Krakow, Poland, 30 501
        • Site PL48001
      • Szczecin, Poland, 70-111
        • Site PL48013
      • Warszawa, Poland, 00 507
        • Site PL48005
      • Wroclaw, Poland, 50-556
        • Site PL48006
      • Wroclaw, Poland, 51 124
        • Site PL48009
      • Zamosc, Poland, 20-400
        • Site PL48014
      • Almada, Portugal, 2800-455
        • Site PT35121
      • Aveiro, Portugal, 3800-266
        • Site PT35127
      • Cascais, Portugal, 2750-663
        • Site PT35139
      • Faro, Portugal, 8005-546
        • Site PT35117
      • Gaeiras, Portugal, 2510-702
        • Site PT35128
      • Leiria, Portugal, 2400-441
        • Site PT35114
      • Porto, Portugal, 4099-001
        • Site PT35102
      • Setubal, Portugal, 2900-655
        • Site PT35122
      • Bucharest, Romania, 011794
        • Site RO40018
      • Bucharest, Romania
        • Site RO40015
      • Bucharest, Romania
        • Site RO40019
      • Bucuresti, Romania, 22328
        • Site RO40003
      • Oradea, Romania, 410562
        • Site RO40004
      • Kaluga, Russian Federation, 248007
        • Site RU70008
      • Moscow, Russian Federation, 119992
        • Site RU70051
      • Moscow, Russian Federation, 125284
        • Site RU70005
      • Nizhny Novgorod, Russian Federation, 603032
        • Site RU70003
      • Omsk, Russian Federation, 644112
        • Site RU70004
      • Rostov-on-Don, Russian Federation, 344029
        • Site RU70014
      • Saint Petersburg, Russian Federation, 197089
        • Site RU70002
      • Saint Petersburg, Russian Federation, 190103
        • Site RU70072
      • Saint-Petersburg, Russian Federation, 196247
        • Site RU70011
      • Saint-Petersburg, Russian Federation, 197374
        • Site RU70050
      • Sankt-Peterburg, Russian Federation, 197110
        • Site RU70030
      • Smolensk, Russian Federation, 214006
        • Site RU70006
      • Volgograd, Russian Federation, 404120
        • Site RU70037
      • Yaroslavl, Russian Federation, 150062
        • Site RU70001
      • Belgrade, Serbia, 11000
        • Site RS38102
      • Belgrade, Serbia, 11000
        • Site RS38105
      • Belgrade, Serbia
        • Site RS38104
      • Belgrade, Serbia, 11000
        • Site RS38120
      • Krusevac, Serbia, 37000
        • Site RS38117
      • Nis, Serbia
        • Site RS38101
      • Zrenjanin, Serbia
        • Site RS38116
      • Koshice, Slovakia, 04001
        • Site SK42102
      • Levice, Slovakia, 93401
        • Site SK42119
      • Lučenec, Slovakia, 984 01
        • Site SK42120
      • Puchov, Slovakia, 020 01
        • Site SK42113
      • Senica, Slovakia, 90501
        • Site SK42116
      • Badalona-Barcelona, Spain, 8916
        • Site ES34002
      • Barcelona, Spain, 08025
        • Site ES34008
      • Barcelona, Spain, 08035
        • Site ES34006
      • Jaen, Spain, 23007
        • Site ES34017
      • Madrid, Spain, 28046
        • Site ES34037
      • Valencia, Spain, 46017
        • Site ES34052
    • A Coruna
      • Santiago de Compostela, A Coruna, Spain, 15706
        • Site ES34041
    • Almeria
      • El Ejido, Almeria, Spain, 04700
        • Site ES34009
    • Madrid
      • Alcorcon, Madrid, Spain, 28922
        • Site ES34010
      • Majadahonda, Madrid, Spain, 28222
        • Site ES34030
    • Vizcaya
      • Galdakao, Vizcaya, Spain, 48960
        • Site ES34011
      • Cambridge, United Kingdom, CB2 0QQ
        • Site GB44008
      • Hull, United Kingdom, HU3 2JZ
        • Site GB44010
      • Leicester, United Kingdom, LE5 4PW
        • Site GB44081
      • Liverpool, United Kingdom, L9 7AL
        • Site GB44079
      • Swansea, United Kingdom, SA6 6NL
        • Site GB44001
    • EastSussex
      • Brighton, EastSussex, United Kingdom, BN2 5BD
        • Site GB44087
    • Kent
      • Canterbury, Kent, United Kingdom, CT1 3NG
        • Site GB44011
    • Staffordshire
      • Stoke on Trent, Staffordshire, United Kingdom, ST4 6QG
        • Site GB44080

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria:

Main Inclusion:

  • Participant is on stable hemodialysis (HD), hemodiafiltration (HDF) or peritoneal dialysis (PD) treatment with the same mode of dialysis for ≥4 months prior to randomization.
  • Participant is on IV or SC epoetin or IV or SC darbepoetin alfa treatment for ≥8 weeks prior to randomization with stable weekly doses (during 4 weeks prior to randomization).
  • Mean of the participant's three most recent Hb values, as measured by central laboratory, during the Screening Period.
  • Participant's alanine aminotransferase (ALT) and aspartate aminotransferase (AST) are ≤3 x upper limit of normal (ULN), and total bilirubin (TBL) is ≤1.5 x ULN

Exclusion Criteria:

Main Exclusion:

  • Participant has received a red blood cell (RBC) transfusion within 8 weeks prior to randomization.
  • Participant has a known hereditary hematologic disease such as thalassemia or sickle cell anemia, pure red cell aplasia, or other known causes for anemia other than Chronic Kidney Disease (CKD).
  • Participant has had a myocardial infarction, acute coronary syndrome, stroke, seizure, or a thrombotic/thrombo-embolic event (e.g., deep vein thrombosis or pulmonary embolism) within 12 weeks prior to randomization.
  • Participant has had uncontrolled hypertension, in the opinion of the investigator, within 2 weeks prior to randomization.
  • Participant has a history of malignancy, except for the following: cancers determined to be cured or in remission for ≥5 years, curatively resected basal cell or squamous cell skin cancers, cervical cancer in situ, or resected colonic polyps.
  • Participant has had any prior organ transplant (that has not been explanted), or participant is scheduled for organ transplantation.

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Roxadustat
Participants received roxadustat three times a week (TIW) for at least 52 weeks up to a maximum of 104 weeks. Participants received initial dose of roxadustat in doses of 100 mg, 150 mg or 200 mg, according to the average weekly dose of epoetin or darbepoetin alfa prior to randomization. Participants' roxadustat dosage was adjusted every 4 weeks to maintain Hb level within the target range 10.0 to 12.0 g/dL. Dose adjustment steps were as follows: 20, 40, 50, 70, 100, 150, 200, 250, 300, and 400 mg. Oral iron treatment of 200 mg was allowed for supplementation to support erythropoiesis. Treatment with intravenous iron was allowed only if certain protocol criteria were met.
Participants received initial dose of roxadustat orally as a tablet in doses of 100 mg, 150 mg or 200 mg, according to the average weekly dose of epoetin or darbepoetin alfa prior to randomization. Participants' roxadustat dosage was adjusted every 4 weeks to maintain Hb level within the target range 10.0 to 12.0 g/dL. Dose adjustment steps were as follows: 20, 40, 50, 70, 100, 150, 200, 250, 300, and 400 mg. Oral iron treatment of 200 mg was allowed for supplementation to support erythropoiesis. Treatment with intravenous iron was allowed only if certain protocol criteria were met.
Other Names:
  • FG-4592
  • ASP1517
Oral iron treatment was recommended for supplementation to support erythropoiesis and as first-line treatment for iron deficiency, unless participant was intolerant to this treatment. For participants receiving roxadustat the recommended daily dose was 200 mg of elemental iron. Participants were advised to take roxadustat at least 1 hour before or 1 hour after oral iron. Intravenous iron supplementation for participants receiving roxadustat was allowed if all of the following criteria were met: The participant's Hb level had not responded adequately to roxadustat following two consecutive dose increases or reached the maximum dose limit, and participant's ferritin was < 100 ng/mL (< 220 pmol/L) or TSAT < 20%, or the participant was intolerant of oral iron therapy. For participants treated with epoetin alfa or darbepoetin alfa, IV iron supplementation was given according to standard of care.
Active Comparator: ESA (Erythropoiesis Stimulating Agent) treatment
Participants received epoetin alfa once weekly, twice weekly or TIW and darbepoetin alfa once a week or once every other week. Participants were treated for at least 52 weeks up to a maximum of 104 weeks. Treatment dosage was adjusted according to the pre-specified rule of keeping the participant's Hb levels between 10.0 to 12.0 g/dL. Participants were not allowed to switch from the epoetin alfa to darbepoetin alfa or vice versa.
Oral iron treatment was recommended for supplementation to support erythropoiesis and as first-line treatment for iron deficiency, unless participant was intolerant to this treatment. For participants receiving roxadustat the recommended daily dose was 200 mg of elemental iron. Participants were advised to take roxadustat at least 1 hour before or 1 hour after oral iron. Intravenous iron supplementation for participants receiving roxadustat was allowed if all of the following criteria were met: The participant's Hb level had not responded adequately to roxadustat following two consecutive dose increases or reached the maximum dose limit, and participant's ferritin was < 100 ng/mL (< 220 pmol/L) or TSAT < 20%, or the participant was intolerant of oral iron therapy. For participants treated with epoetin alfa or darbepoetin alfa, IV iron supplementation was given according to standard of care.
Participants received epoetin alfa via intravenous or subcutaneous injection, once a week, twice a week, or three times a week (TIW). Epoetin alfa dosage was adjusted to maintain Hb level within the target range. Dosing of epoetin alfa was per UK SmPC of Eprex®. Participants received IV iron supplementation according to the standard of care.
Other Names:
  • Eprex
Participants received darbepoetin alfa via intravenous or subcutaneous injection, once a week or once every other week. Darbepoetin alfa dosage was adjusted to maintain Hb level within the target range. Dosing of darbepoetin alfa was per EU SmPC of Aranesp®. Participants received IV iron supplementation according to the standard of care.
Other Names:
  • Aranesp

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Change From Baseline (BL) to the Average Hemoglobin (Hb) in Weeks 28-36 Without Rescue Therapy [EU (EMA)]
Time Frame: Baseline and weeks 28 to 36
Baseline Hb was defined as the mean of four central laboratory Hb values; four latest Hb values prior or on the same date as the first study drug intake. For participants who did not have an available Hb value during the week 28-36 period, imputation rules were applied. For analyses without rescue therapy, participants who used rescue therapy after the initiation of rescue therapy were set to missing for 6 weeks from the start date of rescue therapy. If no Hb value was available, an imputation technique was used, with the mean of all available values from Day 1 to minimum (End of Efficacy Emergent Period) carried forward.
Baseline and weeks 28 to 36
Change From BL to the Average Hb in Weeks 28 to 52 Regardless of Rescue Therapy [US (FDA)]
Time Frame: Baseline and weeks 28 to 52
Baseline Hb was defined as the mean of four central laboratory Hb values: four latest Hb values prior or on the same date as first study drug intake. Change from baseline to the average Hb are observed values. Missing hemoglobin data was imputed for each treatment relying on non-missing data from all participants within each treatment group using the Monte Carlo Markov Chain (MCMC) imputation model with treatment, baseline hemoglobin, randomization stratification factors and the available non missing hemoglobin for each scheduled week.
Baseline and weeks 28 to 52

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Percentage of Participants With Hb Response During Weeks 28 to 36
Time Frame: Weeks 28 to 36
Hb response during weeks 28-36, was defined as mean Hb from 10-12 g/dL without receiving rescue therapy in the 6 weeks prior to, or during, the evaluation period. The percentages and 95% CI were unadjusted, the exact method of Clopper-Pearson was used for 95% CI. The Efficacy Emergent Period was defined as the evaluation period from the Analysis date of first dose intake up to end of treatment (EOT) Visit or last non-missing Hb assessment (for participants who died during the treatment period).
Weeks 28 to 36
Change From BL in Low Density Lipoprotein Cholesterol (LDL-C) to the Average LDL-C of Weeks 12 to 28
Time Frame: Baseline and weeks 12 to 28
Baseline LDL was defined as the LDL value on Day 1. If this value was missing, the latest value prior to first study drug administration was used.
Baseline and weeks 12 to 28
Mean Monthly Intravenous (IV) Iron Use
Time Frame: Day 1 to week 36
Participants with no or missing medication records of IV Iron have their monthly IV Iron use set to 0 mg. For participants who took IV Iron, but without a dosing frequency, the average values were set to missing.
Day 1 to week 36
Change From BL in Short Form-36 (SF-36) Health Survey Physical Functioning (PF) Sub-score to the Average of Weeks 12 to 28
Time Frame: Baseline and weeks 12 to 28
Baseline SF-36 PF was defined as the SF-36 PF value on Day 1.The SF-36 is a Quality of Life (QoL) instrument designed to assess generic health concepts relevant across age, disease, and treatment groups. The SF-36 contains 36 items that measure eight scales: (1) physical functioning (PF); (2) role limitations due to physical health problems (RP); (3) bodily pain (BP); (4) social functioning (SF); (5) general health perceptions (GH); (6) role limitations due to emotional problems (RE); (7) vitality, energy or fatigue (VT); and (8) mental health(MH). Each scale is transformed into 0-100 score, with higher scores indicating better health status. The SF-36 PF consists of 11 questions focused on health and ability to do usual activities, with higher scores indicating better health status.
Baseline and weeks 12 to 28
Change From BL in SF-36 Vitality (VT) Sub-score to the Average of Weeks 12 to 28
Time Frame: Baseline and weeks 12 to 28
Baseline VT Subscore was defined as the VT value on Day 1. The SF-36 is a QoL instrument designed to assess generic health concepts relevant across age, disease, and treatment groups. The SF-36 vitality has four questions with score range from 0-100 with higher scores indicating better vitality status.
Baseline and weeks 12 to 28
Change From BL in Mean Arterial Pressure (MAP) to the Average of Weeks 20 to 28
Time Frame: Baseline and weeks 20 to 28
Baseline MAP was defined as the MAP value on Day 1. If this value was missing, the latest value prior to first study drug administration was used. Mean Arterial Pressure (MAP) is derived as: MAP = (2/3)*DBP + (1/3)*SBP.
Baseline and weeks 20 to 28
Time to First Occurrence of an Increase in Blood Pressure
Time Frame: Weeks 1 to 36
Increase in Blood Pressure was defined as either: Systolic Blood Pressure (SBP) ≥ 170 mmHg and an increase from BL ≥ 20 mmHg, or as: Diastolic Blood Pressure (DBP) ≥ 100 mmHg and an increase from BL ≥ 15 mmHg. For participants who have experienced more than one event, only their first event following study treatment was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Weeks 1 to 36
Change From BL in Mean Arterial Pressure (MAP) to the Average MAP Value of Weeks 20 to 28
Time Frame: Baseline and weeks 20 to 28
Baseline MAP was defined as the MAP value on day 1. If this value was missing, the latest value prior to first study drug administration was used. Mean Arterial Pressure (MAP) is derived as: MAP = (2/3)*DBP + (1/3)*SBP.
Baseline and weeks 20 to 28
Time to First Occurrence of an Increase in Blood Pressure
Time Frame: Weeks 1 to 36
Increase in Blood Pressure was defined as either: SBP ≥ 170 mmHg and an increase from BL ≥ 20 mmHg, or as: DBP ≥ 100 mmHg and an increase from BL ≥ 15 mmHg. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Weeks 1 to 36
Percentage of Participants With a Hb Response During Weeks 28 and 36 Regardless of Use of Rescue Therapy
Time Frame: Weeks 28 to 36
Hb response was defined as mean Hb during weeks 28 to 36 within the target range of 10.0 to 12.0 g/dL. The percentages and 95% CI are unadjusted, the exact method of Clopper-Pearson was used for 95% CI.
Weeks 28 to 36
Change From BL in Hb to Each Postdosing Time Point
Time Frame: Baseline and weeks 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 14, 16, 18,20, 22, 24, 26, 28, 30, 32, 34, 36, 40, 44, 48, 52, 56, 60, 64, 68, 72,76, 80, 84, 88, 92, 96, 100, and 104
Baseline Hb was defined as the mean of four latest central laboratory Hb values prior or on the same date as first study drug intake (pre-dose).
Baseline and weeks 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 14, 16, 18,20, 22, 24, 26, 28, 30, 32, 34, 36, 40, 44, 48, 52, 56, 60, 64, 68, 72,76, 80, 84, 88, 92, 96, 100, and 104
Hb Level Averaged Over Weeks 28 to 36, 44 to 52, and 96 to 104 Without Use of Rescue Therapy
Time Frame: Weeks 28 to 36, 44 to 52, and 96 to 104
Baseline Hb was defined as the mean of four latest central laboratory Hb values prior or on the same date as first study drug intake (pre-dose). Averaged Hb values over weeks 28-36, weeks 44-52 and weeks 96-104 are observed values.
Weeks 28 to 36, 44 to 52, and 96 to 104
Change From BL in Hb to the Average of Weeks 28 to 36, 44 to 52, and 96 to 104 Regardless of the Use of Rescue Therapy
Time Frame: Baseline and weeks 28 to 36, 44 to 52, and 96 to 104
Change from baseline to the average Hb are observed values. Baseline Hb was defined as the mean of four latest central laboratory Hb values prior or on the same date as first study drug intake (pre-dose).
Baseline and weeks 28 to 36, 44 to 52, and 96 to 104
Percentage of Hb Values ≥ 10 g/dL in Weeks 28 to 36, 44 to 52, and 96 to 104 Without Use of Rescue Therapy
Time Frame: Weeks 28-36, 44-52 and 96-104
Percentage for each participant was calculated as Number of Hb values >= 10.0 g/dL / Total number of Hb values*100 in weeks 28 to 36, 44 to 52 and 96 to 104 without use of rescue therapy within 6 weeks prior to and during the 8 week evaluation period.
Weeks 28-36, 44-52 and 96-104
Percentage of Hb Values Within 10.0 to 12.0 g/dL in Weeks 28 to 36, 44 to 52, and 96 to 104 Without Use of Rescue Therapy
Time Frame: Weeks 28-36, 44-52 and 96-104
Percentage for each participant was calculated as Number of Hb values within 10.0-12.0 g/dL / Total number of Hb values*100 in weeks 28 to 36, 44 to 52 and 96 to 104 without use of rescue therapy within 6 weeks prior to and during the 8 week evaluation period.
Weeks 28-36, 44-52 and 96-104
Number of Hospitalizations
Time Frame: Baseline to End of Treatment (EOT) (Up to week 104)
The number of hospitalizations per participant were calculated during the Efficacy Emergent Period. The Efficacy Emergent Period was defined as the evaluation period from the Analysis date of first dose intake up to EOT Visit or last non-missing Hb assessment (for participants who died during the treatment period). It included all Non-Hemodialysis (HD) hospitalizations. The HD days were not counted as hospitalizations, even when performed overnight.
Baseline to End of Treatment (EOT) (Up to week 104)
Number of Days of Hospitalization Per Year
Time Frame: Baseline to EOT (Up to week 104)
The number of days of hospitalizations per year was calculated as the sum of the durations of all non-HD hospitalizations in days (Date of discharge - Date of admission + 1)] / (duration of efficacy emergent period in days / 365.25). In case of missing dates, the hospitalization duration was imputed by the average duration per stay derived from the participants with non-missing duration within the same treatment group.
Baseline to EOT (Up to week 104)
Time to First Hospitalization
Time Frame: Baseline to EOT (Up to week 104)
Time to first hospitalization in years was defined in years as: (First event date during the Efficacy Emergent Period - Analysis date of First dose intake +1)/365.25, and the 'First event date' was defined as 'Date of first Admission and 'Analysis Date of first dose intake. For participants without hospitalization, the time to censoring was calculated as: (Date of End of Efficacy Emergent Period - Analysis Date of first dose intake + 1) / 365.25. Date of End of Efficacy Emergent Period was defined as as the treatment period up to the EOT visit. For participants who have experienced more than one hospitalization, only their first event following study treatment was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Baseline to EOT (Up to week 104)
Time to First Use of Rescue Therapy
Time Frame: Baseline to EOT (Up to week 104)
Rescue therapy was defined as red blood cell (RBC) transfusion for both treatment groups and ESA for roxadustat participants. Only rescue medication that was started during the study treatment and up to end of efficacy emergent period was taken into account and considered as use of rescue medication. For participants who have experienced more than one use of rescue therapy, only their first event following study treatment was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Baseline to EOT (Up to week 104)
Time to First RBC Transfusion
Time Frame: Baseline to EOT (Up to week 104)
For participants who have experienced more than one RBC transfusion, only their first event following study treatment was used. For RBC transfusions, when the number of units was not given but the volume transfused was, the number of units were estimated by volume transfused/250 mL (for transfusion of packed cell units) or volume transfused/500 mL (for transfusion of full blood). Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Baseline to EOT (Up to week 104)
Mean Monthly Number of RBC Packs Per Participant
Time Frame: Baseline to EOT (Up to week 104)
During efficacy emergent period, the mean monthly number of RBC packs was calculated as the sum of blood volume and units transfused between the first dose and up to the last dose in the period divided by duration of efficacy emergent period (in days) divided by 28 days. Participants without medication records of RBC have their number of RBC packs and volume set to 0.
Baseline to EOT (Up to week 104)
Mean Monthly Volume of RBC Transfusion Per Participant
Time Frame: Baseline to EOT (Up to week 104)
During Efficacy Emergent Period, the mean monthly volume of blood transfused was calculated as the sum of blood volume and units transfused between the first dose and up to the last dose in the period divided by duration of efficacy emergent period (in days) divided by 28 days. The Efficacy Emergent Period was defined as the evaluation period from the Analysis date of first dose intake up to EOT Visit or last non-missing Hb assessment (for participants who died during the treatment period).
Baseline to EOT (Up to week 104)
Time to First Use of IV Iron Supplementation
Time Frame: Baseline to EOT (Up to week 104)
For participants who have received more than one IV iron, only their first event following study treatment was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Baseline to EOT (Up to week 104)
Mean Monthly Intravenous (IV) Iron Per Participant During Weeks 37-52 and Weeks 53-104
Time Frame: Weeks 37-52 and weeks 53-104
Participants with no or missing medication records of IV Iron had their monthly IV Iron use set to 0 mg.
Weeks 37-52 and weeks 53-104
Percentage of Participants With Oral Iron Use Only
Time Frame: Baseline to EOT (Up to week 104)
Percentage of participants with/without IV iron only was calculated based on total number of participants within the Efficacy Emergent Period. The Efficacy Emergent Period is defined as the evaluation period from the Analysis date of first dose intake up to EOT Visit or last non-missing Hb assessment (for participants who died during the treatment period).
Baseline to EOT (Up to week 104)
Change From BL to Each Post-dosing Study Visit in Total Cholesterol
Time Frame: Baseline and weeks 8, 28, 52, 104
Baseline assessment was the assessment from Day 1 visit. If baseline value was missing, then the latest screening period value was used as the baseline regardless of fasting status.
Baseline and weeks 8, 28, 52, 104
Change From BL to Each Post-dosing Study Visit in LDL-C/High-density Lipoprotein Cholesterol (HDL-C) Ratio
Time Frame: Baseline and weeks 8, 28, 52, 104
Baseline was defined as the value on Day 1. If baseline value was missing, the latest value prior to first study drug administration was used regardless of fasting.
Baseline and weeks 8, 28, 52, 104
Change From BL to Each Postdosing Study Visit in Non-HDL Cholesterol
Time Frame: Baseline and weeks 8, 28, 52, 104
Baseline was defined as the value on Day 1. If baseline value was missing, the latest value prior to first study drug administration was used regardless of fasting.
Baseline and weeks 8, 28, 52, 104
Change From BL to Each Postdosing Study Visit in Apolipoproteins A1 (ApoA1)
Time Frame: Baseline and weeks 8, 28, 52, 104
Baseline was defined as the value on Day 1. If baseline value was missing, the latest value prior to first study drug administration was used regardless of fasting.
Baseline and weeks 8, 28, 52, 104
Change From BL to Each Postdosing Study Visit in Apolipoproteins B (ApoB)
Time Frame: Baseline and weeks 8, 28, 52, 104
Baseline was defined as the value on Day 1. If baseline value was missing, the latest value prior to first study drug administration was used regardless of fasting.
Baseline and weeks 8, 28, 52, 104
Change From BL to Each Postdosing Study Visit in ApoB/ApoA1 Ratio
Time Frame: Baseline and weeks 8, 28, 52, 104
Baseline was defined as the value on Day 1. If baseline value was missing, the latest value prior to first study drug administration was used.
Baseline and weeks 8, 28, 52, 104
Number of Participants With Mean LDL Cholesterol < 100 mg/dL Over Weeks 12 to 28
Time Frame: Weeks 12 to 28
Missing category for Fasting Only includes non-fasting participants and the participants with missing values.
Weeks 12 to 28
Number of Participants With CKD Who Achieved Antihypertensive Treatment Goal
Time Frame: Weeks 12 to 28
Achieved antihypertensive treatment goal was defined as SBP < 140 mmHg and DBP < 90 mmHg over an evaluation period based on the average of available values in weeks 12-28 (pre-dialysis).
Weeks 12 to 28
Change From BL to the Average of Weeks 12 to 28 in SF-36 Physical Component Score (PCS)
Time Frame: Baseline and weeks 12 to 28
Baseline SF-36 PCS was defined as the SF-36 PCS value on Day 1. SF-36 contains 36-item that measures 8 scales with scores ranging from 0-100: physical functioning (PF); role limitations due to physical health problems (RP); bodily pain (BP); social functioning (SF); general health perceptions (GH); role limitations due to emotional problems (RE); vitality, energy or fatigue (VT); and mental health (MH). These scores are normed to the US population (norm-based scoring had very little impact on results when data was collected in Western European countries) to have a mean of 50 and standard deviation of 10. The PCS was calculated based on all 8 scales and ranges from 5.02-79.78. For each of these above scales, higher scores always indicating better health status.
Baseline and weeks 12 to 28
Change From BL to the Average of Weeks 12 to 28 in Anemia Subscale (AnS) ("Additional Concerns") of Functional Assessment of Cancer Therapy-Anemia (FACT-An) Score
Time Frame: Baseline and weeks 12 to 28
Baseline FACT-An AnS was defined as the FACT-An AnS value on Day 1. Together with the Functional Assessment of Cancer Therapy - General (FACT-G), the Anemia Subscale (AnS) is referred to as the FACT-An Total. The AnS scale contains 13 fatigue specific items (the Fatigue Score) plus 7 items related to anemia. The Anemia AnS score range is 0 to 80. For the above score, a higher score indicates better QoL.
Baseline and weeks 12 to 28
Change From BL to the Average Value of Weeks 12 to 28 in Total FACT-An Score
Time Frame: Baseline and weeks 12 to 28
Baseline FACT-An Total Score was defined on Day 1. Total Fact-An score is composed of FACT-G and Ans scales. FACT-G contains 27 items that cover four dimensions of well-being: physical (PWB) - 7 items, functional (FWB) - 7 items, social/family (SWB) - 7 items, and emotional (EWB) - 6 items. The AnS scale contains 13 fatigue specific items (the Fatigue Score) plus 7 items related to anemia. The total score is obtained by summation of the scores from PWB, SWB, EWB, FWB and AnS. The FACT-An Total Score scale range is 0-188. A higher score indicates better QoL.
Baseline and weeks 12 to 28
Change From BL to the Average of Weeks 12 to 28 in Euroqol Questionnaire-5 Dimensions 5 Levels (EQ-5D 5L) Visual Analogue Scale (VAS) Score
Time Frame: Baseline and weeks 12 to 28
Baseline assessment was defined as the value on Day 1. The EuroQol Questionnaire -5 Dimensions -5 Levels (EQ-5D-5L) is a self-reported questionnaire, used as a measure of respondents' Health Related Quality of Life (HRQoL) and utility values. The EQ-5D consists of the descriptive system and the visual analogue scale (VAS). The EQ-5D descriptive system comprises 5 dimensions of health: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension has 5 levels: no problems, slight problems, moderate problems, severe problems, extreme problems. The VAS records the respondent's self rated health status on a graduated (0-100) scale, where the endpoints are labeled 'Best imaginable health state' and 'Worst imaginable health state' with higher scores for higher HRQoL.
Baseline and weeks 12 to 28
Percentage of Participants With Improvements Measured by Patients' Global Impression of Change (PGIC)
Time Frame: Baseline and weeks 8, 12, 28, 36, 52, 76, 104
The PGIC is a patient-rated instrument that measures change in participant's overall status on a 7-point scale ranging from 1 (very much improved) to 7 (very much worse), when compared to the start of treatment. The percentage of participants presented includes very much improved, much improved and minimally improved.
Baseline and weeks 8, 12, 28, 36, 52, 76, 104
Change From BL in Serum Hepcidin
Time Frame: Baseline and weeks 4, 12, 20, 36, 52, 104, and End of Study (EOS - up to 108 weeks)
Baseline assessment was assessment from Day 1 visit. If baseline value was missing, the value from screening visit was used. In case of missing data, no imputation rules were applied.
Baseline and weeks 4, 12, 20, 36, 52, 104, and End of Study (EOS - up to 108 weeks)
Change From BL in Serum Ferritin
Time Frame: Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104, and EOS (up to 108 weeks)
Baseline assessment was assessment from Day 1 visit. If baseline value was missing, the value from screening visit was used. In case of missing data, no imputation rules were applied.
Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104, and EOS (up to 108 weeks)
Change From BL in Transferrin Saturation (TSAT)
Time Frame: Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104 and EOS (up to 108 weeks)
Baseline assessment was assessment from Day 1 visit. If baseline value was missing, the value from screening visit was used. In case of missing data, no imputation rules were applied.
Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104 and EOS (up to 108 weeks)
Change From BL in Glycated Hemoglobin (HbA1c) Level to Weeks 12, 28, 36, 44, 52, 60, 84, 104 and EOS (up to Week 108)
Time Frame: Baseline and weeks 12, 28, 36, 44, 52, 60, 84, 104 and EOS (up to 108 weeks)
Percentage of change from baseline to each study visit were calculated for HbA1c. Baseline assessment was assessment from Day 1 visit. If baseline value was missing, the value from screening visit was used. In case of missing data, no imputation rules were applied.
Baseline and weeks 12, 28, 36, 44, 52, 60, 84, 104 and EOS (up to 108 weeks)
Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
Time Frame: Baseline up to EOS (Up to week 108)
Safety was assessed by evaluation of the following variables: (TEAEs; frequency, severity, seriousness, and relationship to study drug), Vital signs (systolic and diastolic blood pressure, pulse, respiratory rate and weight), Clinical laboratory variables (hematology, biochemistry including liver enzymes and total bilirubin, and urinalysis), Physical examination, 12-lead electrocardiogram (ECG) and Vascular Access Thrombosis. All AEs collected during the Safety Emergent Period were counted as TEAE. The TEAE was defined as an adverse event (AE) if it was observed after starting administration of the roxadustat or ESA. Any clinically significant abnormalities were reported as an AEs. All reported deaths after the first study drug administration and up to 28 days after the Analysis Date of Last Dose and considering last dosing frequency.
Baseline up to EOS (Up to week 108)

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Collaborators

Investigators

  • Study Director: Study Physician, Astellas Pharma Europe B.V.

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

November 21, 2014

Primary Completion (Actual)

June 8, 2017

Study Completion (Actual)

July 6, 2018

Study Registration Dates

First Submitted

October 28, 2014

First Submitted That Met QC Criteria

October 28, 2014

First Posted (Estimate)

October 30, 2014

Study Record Updates

Last Update Posted (Actual)

February 4, 2021

Last Update Submitted That Met QC Criteria

January 14, 2021

Last Verified

December 1, 2020

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

Yes

IPD Plan Description

Access to anonymized individual participant level data collected during the study, in addition to study-related supporting documentation, is planned for studies conducted with approved product indications and formulations, as well as compounds terminated during development. Studies conducted with product indications or formulations that remain active in development are assessed after study completion to determine if Individual Participant Data can be shared. Conditions and exceptions are described under the Sponsor Specific Details for Astellas on www.clinicalstudydatarequest.com.

IPD Sharing Time Frame

Access to participant level data is offered to researchers after publication of the primary manuscript (if applicable) and is available as long as Astellas has legal authority to provide the data.

IPD Sharing Access Criteria

Researchers must submit a proposal to conduct a scientifically relevant analysis of the study data. The research proposal is reviewed by an Independent Research Panel. If the proposal is approved, access to the study data is provided in a secure data sharing environment after receipt of a signed Data Sharing Agreement.

IPD Sharing Supporting Information Type

  • Study Protocol
  • Statistical Analysis Plan (SAP)
  • Clinical Study Report (CSR)

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Anemia

Clinical Trials on Roxadustat

3
Subscribe