search
Back to results

Roxadustat in the Treatment of Anemia in Chronic Kidney Disease (CKD) Patients, Not on Dialysis, in Comparison to Darbepoetin Alfa (Dolomites)

Primary Purpose

Anemia in Chronic Kidney Disease in Non-dialysis Patients

Status
Completed
Phase
Phase 3
Locations
International
Study Type
Interventional
Intervention
Roxadustat
Darbepoetin alfa
Sponsored by
Astellas Pharma Europe B.V.
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Anemia in Chronic Kidney Disease in Non-dialysis Patients focused on measuring Non-dialysis, Roxadustat, ASP1517, Chronic Kidney Disease (CKD), Anemia, Hemoglobin

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  • Subject has a diagnosis of CKD, with Kidney Disease Outcomes Quality Initiative (KDOQI) Stage 3, 4 or 5, not on dialysis; with an Estimated Glomerular Filtration Rate (eGFR) <60 mL/min/1.73 m^2 estimated using the abbreviated 4-variable Modification of Diet in Renal Disease (MDRD) equation.
  • The mean of the subject's two most recent (prior to randomization) Hb values during the screening period, obtained at least 4 days apart, must be less than or equal to 10.5 g/dL, with a difference of less than or equal to 1.0 g/dL. The last Hb value must be within 10 days prior to randomization.
  • Subject is deemed suitable for treatment with Erythropoiesis Stimulating Agent (ESA) using the criteria specified in the Kidney Disease Improving Global Outcomes (KDIGO) 2012 recommendation considering the rate of fall of Hb concentration, prior response to iron therapy, the risk of needing a transfusion, the risks related to ESA therapy and the presence of symptoms attributable to anemia.
  • Subject has a serum folate level greater than or equal to lower limit of normal (LLN) at screening.
  • Subject has a serum vitamin B12 level greater than or equal to LLN at screening.
  • Subject's alanine aminotransferase (ALT) and aspartate aminotransferase (AST) are less than or equal to 3 x upper limit of normal (ULN), and total bilirubin (TBL) is less than or equal to 1.5 x ULN.
  • Subject's body weight is 45.0 kg to a maximum of 160.0 kg.
  • Male subject must not donate sperm starting from screening, throughout the study period and up to 12 weeks after final study drug administration.

Exclusion Criteria:

  • Subject has received any Erythropoiesis Stimulating Agent (ESA) treatment within 12 weeks prior to randomization.
  • Subject has received any dose of IV iron within 6 weeks prior to randomization.
  • Subject has received a Red Blood Cell (RBC) transfusion within 8 weeks prior to randomization.
  • Subject has a known history of myelodysplastic syndrome or multiple myeloma.
  • Subject 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).
  • Subject has a known hemosiderosis, hemochromatosis, coagulation disorder, or hypercoagulable condition.
  • Subject has a known chronic inflammatory disease that could impact erythropoiesis (e.g., systemic lupus erythematosus, rheumatoid arthritis, celiac disease) even if it is currently in remission.
  • Subject is anticipated to undergo elective surgery that is expected to lead to significant blood loss during the study period or anticipated elective coronary revascularization.
  • Subject has active or chronic gastrointestinal bleeding.
  • Subject has received any prior treatment with roxadustat or a Hypoxia-inducible factor prolyl hydroxylase inhibitor (HIF-PHI).
  • Subject has been treated with iron-chelating agents within 4 weeks prior to randomization.
  • Subject has a history of chronic liver disease (e.g., cirrhosis or fibrosis of the liver).
  • Subject has known New York Heart Association Class III or IV congestive heart failure.
  • Subject has had a myocardial infarction, acute coronary syndrome, stroke, seizure, or a thrombotic/thromboembolic event (e.g., deep vein thrombosis or pulmonary embolism) within 12 weeks prior to randomization.
  • Subject has one or more contraindications for treatment with darbepoetin alfa:

    • Uncontrolled hypertension, or two or more blood pressure values of SBP greater than or equal to 160 mmHg or DBP greater than or equal to 95 mmHg (within 2 weeks prior to randomization).
    • Known hypersensitivity to darbepoetin alfa, recombinant human erythropoietin, or any of the excipients.
  • Subject has a diagnosis or suspicion (e.g., complex kidney cyst of Bosniak Category 2F or higher) of renal cell carcinoma as shown on renal ultrasound within 12 weeks prior to randomization.
  • Subject has a history of malignancy, except for the following: cancers determined to be cured or in remission for greater than or equal to 5 years, curatively resected basal cell or squamous cell skin cancers, cervical cancer in situ, or resected colonic polyps.
  • Subject is positive for any of the following:

    • human immunodeficiency virus (HIV).
    • hepatitis B surface antigen (HBsAg).
    • or anti-hepatitis C virus antibody (anti-HCV Ab).
  • Subject has an active clinically significant infection that is manifested by White Blood Count (WBC) > Upper Limit of Normal (ULN), and/or fever, in conjunction with clinical signs or symptoms of infection within one week prior to randomization.
  • Subject has a known untreated proliferative diabetic retinopathy, diabetic macular edema, macular degeneration or retinal vein occlusion.
  • Subject has had any prior organ transplant (that has not been explanted), subject is scheduled for organ transplantation, or subject is likely to initiate renal replacement therapy including dialysis within the first year of the study.
  • Subject will be excluded from participation if any of the following apply:

    • subject has received investigational therapy within 30 days or 5 half lives or limit set by national law, whichever is longer, prior to initiation of screening, or
    • any condition which makes the subject unsuitable for study participation.
  • Subject has an anticipated use of dapsone in any dose amount or chronic use of acetaminophen/paracetamol >2.0 g/day during the treatment or follow-up period of the study.
  • Subject has a history of alcohol or drug abuse within 2 years prior to randomization

Sites / Locations

  • Site AT43009
  • Site BY37503
  • Site BY37501
  • Site BY37506
  • Site BY37507
  • Site BY37505
  • Site BY37502
  • Site BG35907
  • Site BG35927
  • Site BG35916
  • Site HR38505
  • Site HR38504
  • Site HR38510
  • Site HR38502
  • Site HR38509
  • Site CZ42018
  • Site CZ42008
  • Site CZ42021
  • Site FI35810
  • Site FR33004
  • Site FR33009
  • Site FR33010
  • Site FR33062
  • Site FR33012
  • Site FR33007
  • Site GE99503
  • Site GE99504
  • Site GE99502
  • Site DE49073
  • Site DE49054
  • Site DE49065
  • Site DE49075
  • Site DE49057
  • Site HU36028
  • Site HU36029
  • Site HU36027
  • Site HU36008
  • Site HU36046
  • Site IE35301
  • Site IL97202
  • Site IL97215
  • Site LV37101
  • Site LV37104
  • Site ME38202
  • Site ME38201
  • Site NL31005
  • Site MK38901
  • Site MK38903
  • Site PL48001
  • Site PL48066
  • Site PL48013
  • Site PL48007
  • Site PL48004
  • Site PL48009
  • Site PL48059
  • Site PT35120
  • Site PT35131
  • Site PT35112
  • Site PT35119
  • Site PT35118
  • Site PT35133
  • Site PT35122
  • Site PT35132
  • Site RO40003
  • Site RO40021
  • Site RO40012
  • Site RO40004
  • Site RU70024
  • Site RU70054
  • Site RU70047
  • Site RU70006
  • Site RU70003
  • Site RU70004
  • Site RU70014
  • Site RU70011
  • Site RU70002
  • Site RU70060
  • Site RU70057
  • Site RU70001
  • Site RS38102
  • Site RS38105
  • Site RS38103
  • Site RS38104
  • Site RS38117
  • Site RS38101
  • Site SK42109
  • Site SK42102
  • Site SK42113
  • Site SK42116
  • Site SI38615
  • Site SI38603
  • Site SI38619
  • Site SI38609
  • Site ES34049
  • Site ES34026
  • Site ES34039
  • Site ES34054
  • Site ES34017
  • Site ES34037
  • Site ES34010
  • Site ES34030
  • Site ES34041
  • Site UA38009
  • Site UA38021
  • Site UA38006
  • Site UA38016
  • Site UA38011
  • Site UA38017
  • Site UA38007
  • Site UA38008
  • Site UA38001
  • Site UA38018
  • Site GB44098
  • Site GB44064
  • Site GB44099
  • Site GB44102
  • Site GB44081
  • Site GB44086
  • Site GB44006
  • Site GB44082
  • Site GB44097
  • Site GB44100
  • Site GB44101
  • Site GB44080
  • Site GB44001

Arms of the Study

Arm 1

Arm 2

Arm Type

Experimental

Active Comparator

Arm Label

Roxadustat

Darbepoetin alfa

Arm Description

Participants received roxadustat orally according to the tiered weight-based approach, with starting dose of 70 mg (milligram) given thrice weekly (TIW) to participants weighing up to 70 kg (kilogram) and 100 mg given TIW to participants weighing more than 70 kg. Dose-titration was performed based upon regular measurement of Hb levels until participants achieved central Hb value of ≥ 11.0 g/dL (gram per deciliter) and Hb increase from baseline of ≥ 1.0 g/dL at two consecutive study visits, separated by at least 5 days. Once target Hb level was reached participants entered maintenance period during which roxadustat dosage was adjusted every 4 weeks to maintain participants Hb level within the target range of 10.0 g/dL and 12.0 g/dL. Participants received roxadustat up to a maximum of 104 weeks.

Participants received initial dose of darbepoetin alfa based upon the weight (either 0.45 μg/kg (microgram per kilogram), as a single subcutaneous or intravenous (IV) injection once weekly or 0.75 μg/kg, as a single subcutaneous injection once every 2 weeks) as per European Summary of Product Characteristics (EU SmPC) along with IV iron supplementation according to the standard of care. Dose-adjustment was performed based upon regular measurement of Hb levels until participants achieved central Hb value of ≥ 11.0 g/dL and Hb increase from baseline of ≥ 1.0 g/dL at two consecutive study visits, separated by at least 5 days. Once target Hb level was reached participants entered maintenance period during which darbepoetin alfa dosage was adjusted every 4 weeks to maintain participants Hb level within the target range of 10.0 g/dL and 12.0 g/dL. Participants received darbepoetin alfa for up to a maximum of 104 weeks.

Outcomes

Primary Outcome Measures

Percentage of Participants With a Hb Response to Treatment at Two Consecutive Visits During the First 24 Weeks of Treatment Without Rescue Therapy
Hb response was measured as Yes or No. Response Yes (responders) was defined as: Hb ≥11.0 g/dL and Hb change from baseline by ≥ 1.0 g/dL, for participants with baseline Hb > 8.0 g/dL; or Hb change from baseline by ≥ 2.0 g/dL, for participants with baseline Hb ≤ 8.0 g/dL at two consecutive visits with available data separated at least 5 days during the first 24 weeks of treatment without having received rescue therapy (red blood cell [RBC] transfusion for all participants or darbepoetin for roxadustat treated participant).

Secondary Outcome Measures

Change From Baseline in Hb to the Average Hb of Weeks 28 to 36 Without Rescue Therapy Within 6 Weeks Prior to and During This 8-Week Evaluation Period
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (predose).
Change From Baseline in Low Density Lipoprotein Cholesterol (LDL-C) to the Average LDL-C of Weeks 12 to 28
Baseline LDL-C was defined as the LDL-C value on day 1. If this value was missing, the latest value prior to first study drug administration was used.
Time to First Intravenous Iron Use
Participants were analyzed 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 end of treatment (EOT) Visit or last non-missing Hb assessment (for participants who died during the treatment period). For participants who had received more than one intravenous 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. Percentage of participants were reported in this outcome measure.
Change From Baseline in Short Form-36 (SF-36) Physical Functioning (PF) Sub-Score to the Average PF Sub Score in 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 measured eight scales: (1) physical functioning; (2) role limitations due to physical health problems; (3) bodily pain; (4) social functioning; (5) general health perceptions; (6) role limitations due to emotional problems; (7) vitality, energy or fatigue; and (8) mental health. Each scale is transformed into 0-100 score, with higher scores indicating better health status. The SF-36 PF consisted of 11 questions that focused on health and ability to do usual activities, with higher scores indicating better health status.
Change From Baseline in SF-36 Vitality (VT) Sub-Score to the Average VT Sub-Score in 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.
Change From Baseline in Mean Arterial Pressure (MAP) to the Average MAP Value in Weeks 20 to 28: Per Protocol Set
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. MAP was derived as: MAP = (2/3)*diastolic blood pressure (DBP) + (1/3)*systolic blood pressure (SBP).
Time to First Occurrence of Hypertension During Weeks 1 to 36: Per Protocol Set
Hypertension was defined as either SBP ≥ 170 mmHg and an increase from baseline ≥ 20 mmHg or as DBP ≥ 110 mmHg and an increase from baseline ≥ 15 mmHg. For participants who had 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. Percentage of participants were reported in this outcome measure.
Change From Baseline in MAP to the Average MAP Value in Weeks 20 to 28: Full Analysis Set
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. MAP was derived as: MAP = (2/3)*DBP + (1/3)*SBP.
Time to First Occurrence of Hypertension During Weeks 1 to 36: Full Analysis Set
Hypertension was defined as either SBP ≥ 170 mmHg and an increase from baseline ≥ 20 mmHg or as DBP ≥ 110 mmHg and an increase from baseline ≥ 15 mmHg. For participants who had 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. Percentage of participants were reported in this outcome measure.
Change From Baseline in Hb to the Average Hb Value of Weeks 28 to 52 Regardless of Rescue Therapy
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (pre-dose).
Time to First Hb Response During First 24 Weeks of Treatment Regardless of Administration of Rescue Therapy
Participants were analyzed 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). For participants who had experienced more than one event, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion. Percentage of participants were reported in this outcome measure.
Time to First Hb Response During First 24 Weeks of Treatment Without Rescue Therapy
Participants were analyzed 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). For participants who had experienced more than one event, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion. Percentage of participants were reported in this outcome measure.
Hb Level Averaged Over Weeks 28 to 36, 44 to 52, 72 to 80 and 96 to 104 Without Rescue Therapy
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (predose).
Change From Baseline in Hb to Each Postdosing Time Point Regardless Use of Rescue Therapy
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (pre-dose).
Change From Baseline in Hb to Average Hb Value of Weeks 28 to 36, 44 to 52, 72 to 80, 96 to 104 Regardless of Use of Rescue Therapy
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (predose).
Percentage of Hb Values >=10 g/dL and 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
Percentage for each participant was calculated from the 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.
Time to First Hb Rate of Rise > 2 g/dL Within 4 Weeks
Participants were analyzed 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). For participants who had experienced more than one event, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Number of Hospitalizations
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).
Number of Days of Hospitalization Per Year
The number of days of hospitalization per year was calculated as the sum of the durations of all hospitalizations in days (minimum [date of discharge, end of efficacy of emergent period] - date of admission + 1) / (duration of efficacy emergent period in days / 365.25). 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).
Time to First Hospitalization
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. 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. Percentage of participants were reported in this outcome measure.
Time to First Use of RBC Transfusion
Participants were analyzed 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). For participants who had experienced more than one RBC transfusion, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Number of RBC Packs
The number of RBC packs were calculated as the sum of units transfused 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). Participants with no medication records of RBC have their number of RBC packs set to 0.
Volume of RBC Transfused
The volume of blood transfused was calculated as the sum of blood volume transfused 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). Participants with no medication records of RBC have their volume set to 0.
Number of Particpants Who Received RBC Transfusions
Participants who received RBC transfusions during the efficacy emergent period were reported. 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).
Time to First Use of Rescue Therapy
Rescue therapy for participants in the roxadustat group included RBC transfusion or ESA therapy and for participants in the darbepoetin alfa group included RBC transfusion only. Participants were analyzed 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). For participants who have experienced more than one use of rescue therapy (i.e. RBC and ESA), only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Number of Participants Who Received Rescue Therapy (Composite of RBC Transfusions (All Participants) and Darbepoetin Alfa Use (Roxadustat Treated Participants Only)
Rescue therapy for participants in the roxadustat group included RBC transfusion or ESA therapy and for participants in the darbepoetin alfa group included RBC transfusion only. Participants were analyzed 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). For participants who have experienced more than one use of rescue therapy (i.e. RBC and ESA), only their first event was used.
Mean Monthly Intravenous Iron Per Participant During Weeks 37 to 52 and 53 to 104
Participants with no or missing medication records of IV Iron had their monthly IV Iron use set to 0 mg.
Time to First Use of IV Iron Supplementation
Participants were analyzed 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). For participants who had 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. Percentage of participants were reported in this outcome measure.
Percentage of Participants With Oral Iron Use Only
Percentage of participants with oral iron use only were calculated based on total number of participants within 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).
Change From Baseline to Weeks 8, 28, 52 and 104 in Total Cholesterol
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.
Change From Baseline to Weeks 8, 28, 52 and 104 in LDL-C/High-Density Lipoprotein Cholesterol (HDL-C) Ratio
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.
Change From Baseline to Weeks 8, 28, 52 and 104 in Non-HDL Cholesterol
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.
Change From Baseline to Weeks 8, 28, 52 and 104 in Apolipoproteins A1 (ApoA1)
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.
Change From Baseline to Weeks 8, 28, 52 and 104 in Apolipoproteins B (ApoB)
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.
Change From Baseline to Weeks 8, 28, 52 and 104 in ApoB/ApoA1 Ratio
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.
Number of Participants With Mean LDL Cholesterol < 100 mg/dL
Missing category for fasting only includes non-fasting participants and the participants with missing values.
Number of Participants Who Had Achieved Antihypertensive Treatment Goal
Achieved antihypertensive treatment goal was defined as SBP < 130 mmHg and DBP < 80 mmHg over an evaluation period defined as the average of available values in weeks 12 to 28 and 36 to 52.
Change From Baseline to the Average of Weeks 12 to 28 and 36 to 52 in SF-36 Physical Component Score (PCS)
Baseline SF-36 PCS was defined as the SF-36 PCS 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 contains 36 items that measured eight scales: (1) physical functioning; (2) role limitations due to physical health problems; (3) bodily pain; (4) social functioning; (5) general health perceptions; (6) role limitations due to emotional problems; (7) vitality, energy or fatigue; and (8) mental health. Each scale is transformed into 0-100 score, with higher scores indicating better health status. The PCS was calculated based on all 8 scales and ranged from 5.02-79.78. For each of these above scales, higher scores always indicated better health status.
Change From Baseline to the Average of Weeks 12 to 28 and 36 to 52 in Anemia Subscale (AnS) (Additional Concerns) of Functional Assessment of Cancer Therapy-Anemia (FACT-An) Score
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 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. Higher scores indicated better QoL.
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in FACT-An Total Score
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 well being (PWB) - 7 items, functional well being (FWB) - 7 items, social/family well being (SWB) - 7 items, and emotional well being (EWB) - 6 items. The AnS scale contains 13 fatigue specific items (the Fatigue Score) plus 7 items related to anemia. The total score was obtained by summation of the scores from PWB, SWB, EWB, FWB and AnS. The FACT-An Total Score scale range was 0-188. A higher score indicated better QoL.
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in FACT-An Trial Outcome Index (TOI) Score
Baseline FACT-An total TOI Score was defined on day 1. Total FACT-An TOI score is a sum of PWB subscale score, FWB subscale score and Ans scale score. Fact-An TOI scale contains 14 items that cover four dimensions of well-being: PWB -7 items, FWB -7 items, where score range for each PWB subscale and FWB subscale is 0-28. The AnS scale contains 13 fatigue specific items (the Fatigue Score) plus 7 items related to anemia, where score range for Ans scale is 0-80. The total score was obtained by summation of the scores from PWB, FWB and AnS. The FACT-An Total TOI score range was 0-136. A higher score indicated better QoL.
Change From Baseline to the Average Value of Weeks 12 to 28 in Euroqol Questionnaire-5 Dimensions 5 Levels (EQ-5D 5L) Visual Analogue Scale (VAS) Score
Baseline assessment was defined as the value on day 1. The 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 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.
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in Work Productivity and Activity Impairment-Anemic Symptoms (WPAI:ANS) Score: Percent Work Time Missed
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Multiply scores by 100 to express in percentages. Percent work time missed due to problem: Q2/(Q2+Q4).
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in WPAI:ANS Score: Percent Impairment While Working
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Percent impairment while working due to problem: Q5/10.
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in WPAI:ANS Score: Percent Overall Work Impairment
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Percent overall work impairment due to problem: Q2/(Q2+Q4)+[(1-(Q2/(Q2+Q4))x(Q5/10)].
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in WPAI:ANS Score: Percent Activity Impairment
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Percent activity impairment due to problem: Q6/10.
Percentage of Participants With Improvements Measured by Patients' Global Impression of Change (PGIC)
The PGIC is a patient-rated instrument that measured 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 included very much improved, much improved and minimally improved.
Change From Baseline to Each Scheduled Measurement in Serum Ferritin
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.
Change From Baseline to Each Scheduled Measurement in Transferrin Saturation (TSAT)
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.
Change From Baseline to Each Scheduled Measurement in Glycated Hemoglobin (HbA1c)
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.
Change From Baseline to Each Scheduled Measurement in Fasting Blood Glucose
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.
Change From Baseline to Each Scheduled Measurement in Estimated Glomerular Filtration Rate (eGFR)
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.
Rate of Progression of Chronic Kidney Disease Measured by eGFR Slope Over Time
Annualized eGFR slope over time was estimated by a random slopes and intercepts model using all available eGFR values (one baseline and all post-treatment values up to EOT period or start of dialysis adjusted on baseline Hb, region, CV history at baseline and the interaction terms (baseline eGFR by timepoint and baseline Hb by timepoint). All assessments collected after initiation of dialysis (acute or chronic) were excluded from the analysis. Baseline assessment was the assessment from day 1 visit. If this value was missing, the value from screening visit was used.
Change From Baseline to Each Scheduled Measurement in Urine Albumin/Creatinine Ratio (UACR)
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.
Time to Doubling of Serum Creatinine or Chronic Dialysis or Renal Transplant Compared to Baseline
For participants who had doubled their serum creatinine or had chronic dialysis or renal transplant more than once, only their first occurrence during safety emergent period was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Number of Participants With End Stage Renal Disease (ESRD)
Occurrence of end stage renal disease during the study (i.e from day 1 up to the end of study) was defined as at least one of the following: underwent >30 days dialysis therapy, received kidney transplant, planned kidney transplant, physician recommended renal replacement therapy and participant refused therapy, began dialysis and died < 30 days later.
Time to Chronic Kidney Disease Progression (Composite of Doubling Serum Creatinine, Chronic Dialysis or Renal Transplant, and Death)
Chronic kidney disease progression was defined as date of occurrence of chronic dialysis or date of renal transplant or doubled serum creatinine or date of death, whichever came first. For participants who had chronic dialysis or renal transplant or died, only their first occurrence during the safety emergent period was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time to Chronic Dialysis or Renal Transplant or Death
For participants who had chronic dialysis or renal transplant or died, only their first occurrence during the safety emergent period was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time to at Least 40% Decrease in eGFR From Baseline, Chronic Dialysis or Renal Transplant
For participants who had at least 40% decrease in eGFR from baseline, chronic dialysis or renal transplant during the safety emergent period, only their first occurrence was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by kaplan-meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Number of Participants With Treatment Emergent Adverse Events (TEAEs)
An AE was defined as any untoward medical occurrence in a participant who was given the study drug or who had undergone study procedures and did not necessarily have a causal relationship with this treatment. All AEs collected during the safety emergent period were counted as TEAE. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Based on national cancer institute common terminology criteria (NCI-CTCAE), AEs were graded as grade 1=mild, grade 2=moderate, grade 3 =severe or medically significant, grade 4 =life threatening, grade 5 =death related to AE. All reported deaths after the first study drug administration and up to 28 days after the analysis date of last dose were based on last dosing frequency.

Full Information

First Posted
December 20, 2013
Last Updated
April 19, 2021
Sponsor
Astellas Pharma Europe B.V.
Collaborators
FibroGen
search

1. Study Identification

Unique Protocol Identification Number
NCT02021318
Brief Title
Roxadustat in the Treatment of Anemia in Chronic Kidney Disease (CKD) Patients, Not on Dialysis, in Comparison to Darbepoetin Alfa
Acronym
Dolomites
Official Title
A Phase 3, Randomized, Open-Label, Active-Controlled Study to Evaluate the Efficacy and Safety of Roxadustat in the Treatment of Anemia in Chronic Kidney Disease Patients Not on Dialysis
Study Type
Interventional

2. Study Status

Record Verification Date
April 2021
Overall Recruitment Status
Completed
Study Start Date
March 12, 2014 (Actual)
Primary Completion Date
March 23, 2018 (Actual)
Study Completion Date
November 6, 2019 (Actual)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Astellas Pharma Europe B.V.
Collaborators
FibroGen

4. Oversight

Data Monitoring Committee
Yes

5. Study Description

Brief Summary
The primary objective of this study was to evaluate the efficacy of roxadustat compared to darbepoetin alfa in the treatment of anemia in nondialysis-dependent chronic kidney disease (NDD CKD) participants.
Detailed Description
This was a phase 3, multicenter, randomized, open-label, active-controlled study. The study was planned to provide key efficacy and safety data for the approval of roxadustat in the treatment of anemia associated with CKD. Participants assigned to roxadustat treatment were administered roxadustat orally as a combination of tablets of different strengths. Participants assigned to darbepoetin alfa treatment were administered darbepoetin alfa subcutaneously or intravenously. The study consisted of 3 study periods: Screening period: up to 6 weeks Treatment period: 104 weeks Follow-up period: 4 weeks until planned study end (end of year 2)

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Anemia in Chronic Kidney Disease in Non-dialysis Patients
Keywords
Non-dialysis, Roxadustat, ASP1517, Chronic Kidney Disease (CKD), Anemia, Hemoglobin

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 3
Interventional Study Model
Parallel Assignment
Masking
None (Open Label)
Allocation
Randomized
Enrollment
616 (Actual)

8. Arms, Groups, and Interventions

Arm Title
Roxadustat
Arm Type
Experimental
Arm Description
Participants received roxadustat orally according to the tiered weight-based approach, with starting dose of 70 mg (milligram) given thrice weekly (TIW) to participants weighing up to 70 kg (kilogram) and 100 mg given TIW to participants weighing more than 70 kg. Dose-titration was performed based upon regular measurement of Hb levels until participants achieved central Hb value of ≥ 11.0 g/dL (gram per deciliter) and Hb increase from baseline of ≥ 1.0 g/dL at two consecutive study visits, separated by at least 5 days. Once target Hb level was reached participants entered maintenance period during which roxadustat dosage was adjusted every 4 weeks to maintain participants Hb level within the target range of 10.0 g/dL and 12.0 g/dL. Participants received roxadustat up to a maximum of 104 weeks.
Arm Title
Darbepoetin alfa
Arm Type
Active Comparator
Arm Description
Participants received initial dose of darbepoetin alfa based upon the weight (either 0.45 μg/kg (microgram per kilogram), as a single subcutaneous or intravenous (IV) injection once weekly or 0.75 μg/kg, as a single subcutaneous injection once every 2 weeks) as per European Summary of Product Characteristics (EU SmPC) along with IV iron supplementation according to the standard of care. Dose-adjustment was performed based upon regular measurement of Hb levels until participants achieved central Hb value of ≥ 11.0 g/dL and Hb increase from baseline of ≥ 1.0 g/dL at two consecutive study visits, separated by at least 5 days. Once target Hb level was reached participants entered maintenance period during which darbepoetin alfa dosage was adjusted every 4 weeks to maintain participants Hb level within the target range of 10.0 g/dL and 12.0 g/dL. Participants received darbepoetin alfa for up to a maximum of 104 weeks.
Intervention Type
Drug
Intervention Name(s)
Roxadustat
Other Intervention Name(s)
ASP1517
Intervention Description
Oral tablet.
Intervention Type
Drug
Intervention Name(s)
Darbepoetin alfa
Other Intervention Name(s)
Aranesp
Intervention Description
Subcutaneous or intravenous injection.
Primary Outcome Measure Information:
Title
Percentage of Participants With a Hb Response to Treatment at Two Consecutive Visits During the First 24 Weeks of Treatment Without Rescue Therapy
Description
Hb response was measured as Yes or No. Response Yes (responders) was defined as: Hb ≥11.0 g/dL and Hb change from baseline by ≥ 1.0 g/dL, for participants with baseline Hb > 8.0 g/dL; or Hb change from baseline by ≥ 2.0 g/dL, for participants with baseline Hb ≤ 8.0 g/dL at two consecutive visits with available data separated at least 5 days during the first 24 weeks of treatment without having received rescue therapy (red blood cell [RBC] transfusion for all participants or darbepoetin for roxadustat treated participant).
Time Frame
Baseline to week 24
Secondary Outcome Measure Information:
Title
Change From Baseline in Hb to the Average Hb of Weeks 28 to 36 Without Rescue Therapy Within 6 Weeks Prior to and During This 8-Week Evaluation Period
Description
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (predose).
Time Frame
Baseline and weeks 28 to 36
Title
Change From Baseline in Low Density Lipoprotein Cholesterol (LDL-C) to the Average LDL-C of Weeks 12 to 28
Description
Baseline LDL-C was defined as the LDL-C value on day 1. If this value was missing, the latest value prior to first study drug administration was used.
Time Frame
Baseline and weeks 12 to 28
Title
Time to First Intravenous Iron Use
Description
Participants were analyzed 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 end of treatment (EOT) Visit or last non-missing Hb assessment (for participants who died during the treatment period). For participants who had received more than one intravenous 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. Percentage of participants were reported in this outcome measure.
Time Frame
Weeks 6, 12, 18, 24, 30 and 36
Title
Change From Baseline in Short Form-36 (SF-36) Physical Functioning (PF) Sub-Score to the Average PF Sub Score in Weeks 12 to 28
Description
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 measured eight scales: (1) physical functioning; (2) role limitations due to physical health problems; (3) bodily pain; (4) social functioning; (5) general health perceptions; (6) role limitations due to emotional problems; (7) vitality, energy or fatigue; and (8) mental health. Each scale is transformed into 0-100 score, with higher scores indicating better health status. The SF-36 PF consisted of 11 questions that focused on health and ability to do usual activities, with higher scores indicating better health status.
Time Frame
Baseline and weeks 12 to 28
Title
Change From Baseline in SF-36 Vitality (VT) Sub-Score to the Average VT Sub-Score in Weeks 12 to 28
Description
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.
Time Frame
Baseline and weeks 12 to 28
Title
Change From Baseline in Mean Arterial Pressure (MAP) to the Average MAP Value in Weeks 20 to 28: Per Protocol Set
Description
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. MAP was derived as: MAP = (2/3)*diastolic blood pressure (DBP) + (1/3)*systolic blood pressure (SBP).
Time Frame
Baseline and weeks 20 to 28
Title
Time to First Occurrence of Hypertension During Weeks 1 to 36: Per Protocol Set
Description
Hypertension was defined as either SBP ≥ 170 mmHg and an increase from baseline ≥ 20 mmHg or as DBP ≥ 110 mmHg and an increase from baseline ≥ 15 mmHg. For participants who had 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. Percentage of participants were reported in this outcome measure.
Time Frame
Weeks 1 to 36
Title
Change From Baseline in MAP to the Average MAP Value in Weeks 20 to 28: Full Analysis Set
Description
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. MAP was derived as: MAP = (2/3)*DBP + (1/3)*SBP.
Time Frame
Baseline and weeks 20 to 28
Title
Time to First Occurrence of Hypertension During Weeks 1 to 36: Full Analysis Set
Description
Hypertension was defined as either SBP ≥ 170 mmHg and an increase from baseline ≥ 20 mmHg or as DBP ≥ 110 mmHg and an increase from baseline ≥ 15 mmHg. For participants who had 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. Percentage of participants were reported in this outcome measure.
Time Frame
Weeks 1 to 36
Title
Change From Baseline in Hb to the Average Hb Value of Weeks 28 to 52 Regardless of Rescue Therapy
Description
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (pre-dose).
Time Frame
Baseline and weeks 28 to 52
Title
Time to First Hb Response During First 24 Weeks of Treatment Regardless of Administration of Rescue Therapy
Description
Participants were analyzed 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). For participants who had experienced more than one event, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion. Percentage of participants were reported in this outcome measure.
Time Frame
Weeks 1 to 24
Title
Time to First Hb Response During First 24 Weeks of Treatment Without Rescue Therapy
Description
Participants were analyzed 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). For participants who had experienced more than one event, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion. Percentage of participants were reported in this outcome measure.
Time Frame
Weeks 1 to 24
Title
Hb Level Averaged Over Weeks 28 to 36, 44 to 52, 72 to 80 and 96 to 104 Without Rescue Therapy
Description
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (predose).
Time Frame
Weeks 28 to 36, 44 to 52, 72 to 80 and 96 to 104
Title
Change From Baseline in Hb to Each Postdosing Time Point Regardless Use of Rescue Therapy
Description
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (pre-dose).
Time Frame
Baseline and weeks 1, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 28, 32, 36, 40, 44, 48, 52, 56, 60, 64, 68, 72, 76, 80, 84, 88, 92, 96, 100 and 104
Title
Change From Baseline in Hb to Average Hb Value of Weeks 28 to 36, 44 to 52, 72 to 80, 96 to 104 Regardless of Use of Rescue Therapy
Description
Baseline Hb was defined as the mean of all available central laboratory Hb values collected before or including the date of first study drug intake (predose).
Time Frame
Baseline and weeks 28 to 36, 44 to 52, 72 to 80 and 96 to 104
Title
Percentage of Hb Values >=10 g/dL and 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
Description
Percentage for each participant was calculated from the 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.
Time Frame
Weeks 28 to 36, 44 to 52 and 96 to 104
Title
Time to First Hb Rate of Rise > 2 g/dL Within 4 Weeks
Description
Participants were analyzed 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). For participants who had experienced more than one event, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Number of Hospitalizations
Description
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).
Time Frame
Baseline to EOT (up to week 104)
Title
Number of Days of Hospitalization Per Year
Description
The number of days of hospitalization per year was calculated as the sum of the durations of all hospitalizations in days (minimum [date of discharge, end of efficacy of emergent period] - date of admission + 1) / (duration of efficacy emergent period in days / 365.25). 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).
Time Frame
Baseline to EOT (up to week 104)
Title
Time to First Hospitalization
Description
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. 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. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Time to First Use of RBC Transfusion
Description
Participants were analyzed 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). For participants who had experienced more than one RBC transfusion, only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Number of RBC Packs
Description
The number of RBC packs were calculated as the sum of units transfused 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). Participants with no medication records of RBC have their number of RBC packs set to 0.
Time Frame
Baseline to EOT (up to week 104)
Title
Volume of RBC Transfused
Description
The volume of blood transfused was calculated as the sum of blood volume transfused 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). Participants with no medication records of RBC have their volume set to 0.
Time Frame
Baseline to EOT (up to week 104)
Title
Number of Particpants Who Received RBC Transfusions
Description
Participants who received RBC transfusions during the efficacy emergent period were reported. 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).
Time Frame
Baseline to EOT (up to week 104)
Title
Time to First Use of Rescue Therapy
Description
Rescue therapy for participants in the roxadustat group included RBC transfusion or ESA therapy and for participants in the darbepoetin alfa group included RBC transfusion only. Participants were analyzed 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). For participants who have experienced more than one use of rescue therapy (i.e. RBC and ESA), only their first event was used. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Number of Participants Who Received Rescue Therapy (Composite of RBC Transfusions (All Participants) and Darbepoetin Alfa Use (Roxadustat Treated Participants Only)
Description
Rescue therapy for participants in the roxadustat group included RBC transfusion or ESA therapy and for participants in the darbepoetin alfa group included RBC transfusion only. Participants were analyzed 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). For participants who have experienced more than one use of rescue therapy (i.e. RBC and ESA), only their first event was used.
Time Frame
Baseline to EOT (up to week 104)
Title
Mean Monthly Intravenous Iron Per Participant During Weeks 37 to 52 and 53 to 104
Description
Participants with no or missing medication records of IV Iron had their monthly IV Iron use set to 0 mg.
Time Frame
Weeks 37 to 52 and 53 to 104
Title
Time to First Use of IV Iron Supplementation
Description
Participants were analyzed 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). For participants who had 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. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Percentage of Participants With Oral Iron Use Only
Description
Percentage of participants with oral iron use only were calculated based on total number of participants within 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).
Time Frame
Day 1 to week 36, weeks 37 to 52, weeks 53 to 104, efficacy emergent period (up to week 104)
Title
Change From Baseline to Weeks 8, 28, 52 and 104 in Total Cholesterol
Description
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.
Time Frame
Baseline and weeks 8, 28, 52, 104
Title
Change From Baseline to Weeks 8, 28, 52 and 104 in LDL-C/High-Density Lipoprotein Cholesterol (HDL-C) Ratio
Description
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.
Time Frame
Baseline and weeks 8, 28, 52, 104
Title
Change From Baseline to Weeks 8, 28, 52 and 104 in Non-HDL Cholesterol
Description
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.
Time Frame
Baseline and weeks 8, 28, 52, 104
Title
Change From Baseline to Weeks 8, 28, 52 and 104 in Apolipoproteins A1 (ApoA1)
Description
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.
Time Frame
Baseline and weeks 8, 28, 52, 104
Title
Change From Baseline to Weeks 8, 28, 52 and 104 in Apolipoproteins B (ApoB)
Description
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.
Time Frame
Baseline and weeks 8, 28, 52, 104
Title
Change From Baseline to Weeks 8, 28, 52 and 104 in ApoB/ApoA1 Ratio
Description
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.
Time Frame
Baseline and weeks 8, 28, 52, 104
Title
Number of Participants With Mean LDL Cholesterol < 100 mg/dL
Description
Missing category for fasting only includes non-fasting participants and the participants with missing values.
Time Frame
Weeks 12 to 28 and 36 to 52
Title
Number of Participants Who Had Achieved Antihypertensive Treatment Goal
Description
Achieved antihypertensive treatment goal was defined as SBP < 130 mmHg and DBP < 80 mmHg over an evaluation period defined as the average of available values in weeks 12 to 28 and 36 to 52.
Time Frame
Weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average of Weeks 12 to 28 and 36 to 52 in SF-36 Physical Component Score (PCS)
Description
Baseline SF-36 PCS was defined as the SF-36 PCS 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 contains 36 items that measured eight scales: (1) physical functioning; (2) role limitations due to physical health problems; (3) bodily pain; (4) social functioning; (5) general health perceptions; (6) role limitations due to emotional problems; (7) vitality, energy or fatigue; and (8) mental health. Each scale is transformed into 0-100 score, with higher scores indicating better health status. The PCS was calculated based on all 8 scales and ranged from 5.02-79.78. For each of these above scales, higher scores always indicated better health status.
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average of Weeks 12 to 28 and 36 to 52 in Anemia Subscale (AnS) (Additional Concerns) of Functional Assessment of Cancer Therapy-Anemia (FACT-An) Score
Description
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 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. Higher scores indicated better QoL.
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in FACT-An Total Score
Description
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 well being (PWB) - 7 items, functional well being (FWB) - 7 items, social/family well being (SWB) - 7 items, and emotional well being (EWB) - 6 items. The AnS scale contains 13 fatigue specific items (the Fatigue Score) plus 7 items related to anemia. The total score was obtained by summation of the scores from PWB, SWB, EWB, FWB and AnS. The FACT-An Total Score scale range was 0-188. A higher score indicated better QoL.
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in FACT-An Trial Outcome Index (TOI) Score
Description
Baseline FACT-An total TOI Score was defined on day 1. Total FACT-An TOI score is a sum of PWB subscale score, FWB subscale score and Ans scale score. Fact-An TOI scale contains 14 items that cover four dimensions of well-being: PWB -7 items, FWB -7 items, where score range for each PWB subscale and FWB subscale is 0-28. The AnS scale contains 13 fatigue specific items (the Fatigue Score) plus 7 items related to anemia, where score range for Ans scale is 0-80. The total score was obtained by summation of the scores from PWB, FWB and AnS. The FACT-An Total TOI score range was 0-136. A higher score indicated better QoL.
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average Value of Weeks 12 to 28 in Euroqol Questionnaire-5 Dimensions 5 Levels (EQ-5D 5L) Visual Analogue Scale (VAS) Score
Description
Baseline assessment was defined as the value on day 1. The 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 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.
Time Frame
Baseline and weeks 12 to 28
Title
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in Work Productivity and Activity Impairment-Anemic Symptoms (WPAI:ANS) Score: Percent Work Time Missed
Description
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Multiply scores by 100 to express in percentages. Percent work time missed due to problem: Q2/(Q2+Q4).
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in WPAI:ANS Score: Percent Impairment While Working
Description
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Percent impairment while working due to problem: Q5/10.
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in WPAI:ANS Score: Percent Overall Work Impairment
Description
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Percent overall work impairment due to problem: Q2/(Q2+Q4)+[(1-(Q2/(Q2+Q4))x(Q5/10)].
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Change From Baseline to the Average Value of Weeks 12 to 28 and 36 to 52 in WPAI:ANS Score: Percent Activity Impairment
Description
WPAI consisted of 6 questions (Q1=Employment status; Q2=Hours absent from work due to anaemic symptoms; Q3=Hours absent from work due to other reasons; Q4=Hours actually worked; Q5=Impact of the anaemic symptoms on productivity while working; Q6=Impact of the anaemic symptoms on productivity while doing regular daily activities other than work) and a 1-week recall period. Higher WPAI scores indicated greater activity impairment. Percent activity impairment due to problem: Q6/10.
Time Frame
Baseline, weeks 12 to 28 and 36 to 52
Title
Percentage of Participants With Improvements Measured by Patients' Global Impression of Change (PGIC)
Description
The PGIC is a patient-rated instrument that measured 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 included very much improved, much improved and minimally improved.
Time Frame
Weeks 8, 12, 28, 52, 76, 104, last assessment (week 108)
Title
Change From Baseline to Each Scheduled Measurement in Serum Ferritin
Description
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.
Time Frame
Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104 and end of study (EOS) (up to 108 weeks)
Title
Change From Baseline to Each Scheduled Measurement in Transferrin Saturation (TSAT)
Description
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.
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)
Title
Change From Baseline to Each Scheduled Measurement in Glycated Hemoglobin (HbA1c)
Description
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.
Time Frame
Baseline and weeks 12, 28, 36, 44, 60, 84, 104 and EOS (up to 108 weeks)
Title
Change From Baseline to Each Scheduled Measurement in Fasting Blood Glucose
Description
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.
Time Frame
Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104, 106 and EOS (up to 108 weeks)
Title
Change From Baseline to Each Scheduled Measurement in Estimated Glomerular Filtration Rate (eGFR)
Description
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.
Time Frame
Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104, 106 and EOS (up to 108 weeks)
Title
Rate of Progression of Chronic Kidney Disease Measured by eGFR Slope Over Time
Description
Annualized eGFR slope over time was estimated by a random slopes and intercepts model using all available eGFR values (one baseline and all post-treatment values up to EOT period or start of dialysis adjusted on baseline Hb, region, CV history at baseline and the interaction terms (baseline eGFR by timepoint and baseline Hb by timepoint). All assessments collected after initiation of dialysis (acute or chronic) were excluded from the analysis. Baseline assessment was the assessment from day 1 visit. If this value was missing, the value from screening visit was used.
Time Frame
Baseline up to EOS (up to week 108)
Title
Change From Baseline to Each Scheduled Measurement in Urine Albumin/Creatinine Ratio (UACR)
Description
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.
Time Frame
Baseline and weeks 12, 24, 36, 52, 64, 76, 88 and 104
Title
Time to Doubling of Serum Creatinine or Chronic Dialysis or Renal Transplant Compared to Baseline
Description
For participants who had doubled their serum creatinine or had chronic dialysis or renal transplant more than once, only their first occurrence during safety emergent period was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Number of Participants With End Stage Renal Disease (ESRD)
Description
Occurrence of end stage renal disease during the study (i.e from day 1 up to the end of study) was defined as at least one of the following: underwent >30 days dialysis therapy, received kidney transplant, planned kidney transplant, physician recommended renal replacement therapy and participant refused therapy, began dialysis and died < 30 days later.
Time Frame
Baseline up to EOS (up to week 108)
Title
Time to Chronic Kidney Disease Progression (Composite of Doubling Serum Creatinine, Chronic Dialysis or Renal Transplant, and Death)
Description
Chronic kidney disease progression was defined as date of occurrence of chronic dialysis or date of renal transplant or doubled serum creatinine or date of death, whichever came first. For participants who had chronic dialysis or renal transplant or died, only their first occurrence during the safety emergent period was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Time to Chronic Dialysis or Renal Transplant or Death
Description
For participants who had chronic dialysis or renal transplant or died, only their first occurrence during the safety emergent period was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by Kaplan-Meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula. Percentage of participants were reported in this outcome measure.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Time to at Least 40% Decrease in eGFR From Baseline, Chronic Dialysis or Renal Transplant
Description
For participants who had at least 40% decrease in eGFR from baseline, chronic dialysis or renal transplant during the safety emergent period, only their first occurrence was used. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Data reported was analyzed by kaplan-meier estimate for cumulative proportion and the 95% confidence interval was calculated with Greenwood's formula.
Time Frame
Year 0.5, 1, 1.5 and 2
Title
Number of Participants With Treatment Emergent Adverse Events (TEAEs)
Description
An AE was defined as any untoward medical occurrence in a participant who was given the study drug or who had undergone study procedures and did not necessarily have a causal relationship with this treatment. All AEs collected during the safety emergent period were counted as TEAE. The safety emergent period was defined as the evaluation period from the analysis date of first drug intake up to 28 days after the end of treatment taking into account the different dosing frequencies of the study treatments. Based on national cancer institute common terminology criteria (NCI-CTCAE), AEs were graded as grade 1=mild, grade 2=moderate, grade 3 =severe or medically significant, grade 4 =life threatening, grade 5 =death related to AE. All reported deaths after the first study drug administration and up to 28 days after the analysis date of last dose were based on last dosing frequency.
Time Frame
From first dose of study drug up to end of study (up to week 108)

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Subject has a diagnosis of CKD, with Kidney Disease Outcomes Quality Initiative (KDOQI) Stage 3, 4 or 5, not on dialysis; with an Estimated Glomerular Filtration Rate (eGFR) <60 mL/min/1.73 m^2 estimated using the abbreviated 4-variable Modification of Diet in Renal Disease (MDRD) equation. The mean of the subject's two most recent (prior to randomization) Hb values during the screening period, obtained at least 4 days apart, must be less than or equal to 10.5 g/dL, with a difference of less than or equal to 1.0 g/dL. The last Hb value must be within 10 days prior to randomization. Subject is deemed suitable for treatment with Erythropoiesis Stimulating Agent (ESA) using the criteria specified in the Kidney Disease Improving Global Outcomes (KDIGO) 2012 recommendation considering the rate of fall of Hb concentration, prior response to iron therapy, the risk of needing a transfusion, the risks related to ESA therapy and the presence of symptoms attributable to anemia. Subject has a serum folate level greater than or equal to lower limit of normal (LLN) at screening. Subject has a serum vitamin B12 level greater than or equal to LLN at screening. Subject's alanine aminotransferase (ALT) and aspartate aminotransferase (AST) are less than or equal to 3 x upper limit of normal (ULN), and total bilirubin (TBL) is less than or equal to 1.5 x ULN. Subject's body weight is 45.0 kg to a maximum of 160.0 kg. Male subject must not donate sperm starting from screening, throughout the study period and up to 12 weeks after final study drug administration. Exclusion Criteria: Subject has received any Erythropoiesis Stimulating Agent (ESA) treatment within 12 weeks prior to randomization. Subject has received any dose of IV iron within 6 weeks prior to randomization. Subject has received a Red Blood Cell (RBC) transfusion within 8 weeks prior to randomization. Subject has a known history of myelodysplastic syndrome or multiple myeloma. Subject 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). Subject has a known hemosiderosis, hemochromatosis, coagulation disorder, or hypercoagulable condition. Subject has a known chronic inflammatory disease that could impact erythropoiesis (e.g., systemic lupus erythematosus, rheumatoid arthritis, celiac disease) even if it is currently in remission. Subject is anticipated to undergo elective surgery that is expected to lead to significant blood loss during the study period or anticipated elective coronary revascularization. Subject has active or chronic gastrointestinal bleeding. Subject has received any prior treatment with roxadustat or a Hypoxia-inducible factor prolyl hydroxylase inhibitor (HIF-PHI). Subject has been treated with iron-chelating agents within 4 weeks prior to randomization. Subject has a history of chronic liver disease (e.g., cirrhosis or fibrosis of the liver). Subject has known New York Heart Association Class III or IV congestive heart failure. Subject has had a myocardial infarction, acute coronary syndrome, stroke, seizure, or a thrombotic/thromboembolic event (e.g., deep vein thrombosis or pulmonary embolism) within 12 weeks prior to randomization. Subject has one or more contraindications for treatment with darbepoetin alfa: Uncontrolled hypertension, or two or more blood pressure values of SBP greater than or equal to 160 mmHg or DBP greater than or equal to 95 mmHg (within 2 weeks prior to randomization). Known hypersensitivity to darbepoetin alfa, recombinant human erythropoietin, or any of the excipients. Subject has a diagnosis or suspicion (e.g., complex kidney cyst of Bosniak Category 2F or higher) of renal cell carcinoma as shown on renal ultrasound within 12 weeks prior to randomization. Subject has a history of malignancy, except for the following: cancers determined to be cured or in remission for greater than or equal to 5 years, curatively resected basal cell or squamous cell skin cancers, cervical cancer in situ, or resected colonic polyps. Subject is positive for any of the following: human immunodeficiency virus (HIV). hepatitis B surface antigen (HBsAg). or anti-hepatitis C virus antibody (anti-HCV Ab). Subject has an active clinically significant infection that is manifested by White Blood Count (WBC) > Upper Limit of Normal (ULN), and/or fever, in conjunction with clinical signs or symptoms of infection within one week prior to randomization. Subject has a known untreated proliferative diabetic retinopathy, diabetic macular edema, macular degeneration or retinal vein occlusion. Subject has had any prior organ transplant (that has not been explanted), subject is scheduled for organ transplantation, or subject is likely to initiate renal replacement therapy including dialysis within the first year of the study. Subject will be excluded from participation if any of the following apply: subject has received investigational therapy within 30 days or 5 half lives or limit set by national law, whichever is longer, prior to initiation of screening, or any condition which makes the subject unsuitable for study participation. Subject has an anticipated use of dapsone in any dose amount or chronic use of acetaminophen/paracetamol >2.0 g/day during the treatment or follow-up period of the study. Subject has a history of alcohol or drug abuse within 2 years prior to randomization
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Medical Monitor
Organizational Affiliation
Astellas Pharma Europe B.V.
Official's Role
Study Director
Facility Information:
Facility Name
Site AT43009
City
Vienna
Country
Austria
Facility Name
Site BY37503
City
Brest
ZIP/Postal Code
224027
Country
Belarus
Facility Name
Site BY37501
City
Grodno
ZIP/Postal Code
230017
Country
Belarus
Facility Name
Site BY37506
City
Minsk
ZIP/Postal Code
220036
Country
Belarus
Facility Name
Site BY37507
City
Minsk
ZIP/Postal Code
220116
Country
Belarus
Facility Name
Site BY37505
City
Minsk
ZIP/Postal Code
223040
Country
Belarus
Facility Name
Site BY37502
City
Vitebsk
ZIP/Postal Code
210037
Country
Belarus
Facility Name
Site BG35907
City
Sofia
ZIP/Postal Code
1113
Country
Bulgaria
Facility Name
Site BG35927
City
Sofia
ZIP/Postal Code
1709
Country
Bulgaria
Facility Name
Site BG35916
City
Varna
ZIP/Postal Code
9000
Country
Bulgaria
Facility Name
Site HR38505
City
Karlovac
ZIP/Postal Code
47000
Country
Croatia
Facility Name
Site HR38504
City
Slavonski Brod
ZIP/Postal Code
35000
Country
Croatia
Facility Name
Site HR38510
City
Zagreb
ZIP/Postal Code
10 000
Country
Croatia
Facility Name
Site HR38502
City
Zagreb
ZIP/Postal Code
10000
Country
Croatia
Facility Name
Site HR38509
City
Zagreb
ZIP/Postal Code
10000
Country
Croatia
Facility Name
Site CZ42018
City
Novy Jicin
State/Province
CZ
ZIP/Postal Code
741 01
Country
Czechia
Facility Name
Site CZ42008
City
Liberec
ZIP/Postal Code
46063
Country
Czechia
Facility Name
Site CZ42021
City
Prague
Country
Czechia
Facility Name
Site FI35810
City
Helsinki
ZIP/Postal Code
290
Country
Finland
Facility Name
Site FR33004
City
Avignon
ZIP/Postal Code
84900
Country
France
Facility Name
Site FR33009
City
Colmar
ZIP/Postal Code
68024
Country
France
Facility Name
Site FR33010
City
Grenoble
Country
France
Facility Name
Site FR33062
City
Limoges
ZIP/Postal Code
87042
Country
France
Facility Name
Site FR33012
City
Lyon
ZIP/Postal Code
69437
Country
France
Facility Name
Site FR33007
City
Saint-Priest-en-Jarez
ZIP/Postal Code
42270
Country
France
Facility Name
Site GE99503
City
Tbilisi
ZIP/Postal Code
144
Country
Georgia
Facility Name
Site GE99504
City
Tbilisi
ZIP/Postal Code
144
Country
Georgia
Facility Name
Site GE99502
City
Tbilisi
ZIP/Postal Code
159
Country
Georgia
Facility Name
Site DE49073
City
Cloppenburg
ZIP/Postal Code
49661
Country
Germany
Facility Name
Site DE49054
City
Düsseldorf
ZIP/Postal Code
40210
Country
Germany
Facility Name
Site DE49065
City
Hamburg
ZIP/Postal Code
23397
Country
Germany
Facility Name
Site DE49075
City
Heilbronn
ZIP/Postal Code
74076
Country
Germany
Facility Name
Site DE49057
City
Hoyerswerda
ZIP/Postal Code
02977
Country
Germany
Facility Name
Site HU36028
City
Budapest
ZIP/Postal Code
1097
Country
Hungary
Facility Name
Site HU36029
City
Budapest
Country
Hungary
Facility Name
Site HU36027
City
Kistarcsa
ZIP/Postal Code
2143
Country
Hungary
Facility Name
Site HU36008
City
Pecs
ZIP/Postal Code
H 7624
Country
Hungary
Facility Name
Site HU36046
City
Velence
Country
Hungary
Facility Name
Site IE35301
City
Cork
Country
Ireland
Facility Name
Site IL97202
City
Be'er Ya'akov
ZIP/Postal Code
70300
Country
Israel
Facility Name
Site IL97215
City
Haifa
ZIP/Postal Code
34362
Country
Israel
Facility Name
Site LV37101
City
Riga
ZIP/Postal Code
LV-1002
Country
Latvia
Facility Name
Site LV37104
City
Ventspils
ZIP/Postal Code
LV-3601
Country
Latvia
Facility Name
Site ME38202
City
Niksic
ZIP/Postal Code
81400
Country
Montenegro
Facility Name
Site ME38201
City
Podgorica
ZIP/Postal Code
81000
Country
Montenegro
Facility Name
Site NL31005
City
Utrecht
ZIP/Postal Code
3584 CX
Country
Netherlands
Facility Name
Site MK38901
City
Skopje
ZIP/Postal Code
1000
Country
North Macedonia
Facility Name
Site MK38903
City
Struga
ZIP/Postal Code
6330
Country
North Macedonia
Facility Name
Site PL48001
City
Krakow
ZIP/Postal Code
31-559
Country
Poland
Facility Name
Site PL48066
City
Pulawy
ZIP/Postal Code
24-100
Country
Poland
Facility Name
Site PL48013
City
Szczecin
ZIP/Postal Code
70-111
Country
Poland
Facility Name
Site PL48007
City
Tarnow
ZIP/Postal Code
33-100
Country
Poland
Facility Name
Site PL48004
City
Warszawa
ZIP/Postal Code
04-749
Country
Poland
Facility Name
Site PL48009
City
Wroclaw
Country
Poland
Facility Name
Site PL48059
City
Zamosc
ZIP/Postal Code
22-400
Country
Poland
Facility Name
Site PT35120
City
Almada
ZIP/Postal Code
2801-951
Country
Portugal
Facility Name
Site PT35131
City
Braga
ZIP/Postal Code
4710-243
Country
Portugal
Facility Name
Site PT35112
City
Carnaxide
ZIP/Postal Code
2795-523
Country
Portugal
Facility Name
Site PT35119
City
Evora
ZIP/Postal Code
7000-811
Country
Portugal
Facility Name
Site PT35118
City
Lisboa
ZIP/Postal Code
1069-166
Country
Portugal
Facility Name
Site PT35133
City
Matosinhos
ZIP/Postal Code
4464-513
Country
Portugal
Facility Name
Site PT35122
City
Setubal
ZIP/Postal Code
2910-446
Country
Portugal
Facility Name
Site PT35132
City
Vila Nova de Gaia
ZIP/Postal Code
4434-502
Country
Portugal
Facility Name
Site RO40003
City
Bucharest
ZIP/Postal Code
11234
Country
Romania
Facility Name
Site RO40021
City
Bucharest
ZIP/Postal Code
22328
Country
Romania
Facility Name
Site RO40012
City
Bucuresti
ZIP/Postal Code
10825
Country
Romania
Facility Name
Site RO40004
City
Oradea
ZIP/Postal Code
410469
Country
Romania
Facility Name
Site RU70024
City
Chelyabinsk
ZIP/Postal Code
454047
Country
Russian Federation
Facility Name
Site RU70054
City
Irkutsk
ZIP/Postal Code
664079
Country
Russian Federation
Facility Name
Site RU70047
City
Moscow
ZIP/Postal Code
119992
Country
Russian Federation
Facility Name
Site RU70006
City
Moscow
ZIP/Postal Code
125284
Country
Russian Federation
Facility Name
Site RU70003
City
Nizhny Novgorod
ZIP/Postal Code
603032
Country
Russian Federation
Facility Name
Site RU70004
City
Omsk
ZIP/Postal Code
644112
Country
Russian Federation
Facility Name
Site RU70014
City
Rostov-on-Don
ZIP/Postal Code
344029
Country
Russian Federation
Facility Name
Site RU70011
City
Saint Petersburg
ZIP/Postal Code
196247
Country
Russian Federation
Facility Name
Site RU70002
City
Saint Petersburg
ZIP/Postal Code
197089
Country
Russian Federation
Facility Name
Site RU70060
City
Saratov
ZIP/Postal Code
410039
Country
Russian Federation
Facility Name
Site RU70057
City
Yaroslavl
ZIP/Postal Code
150000
Country
Russian Federation
Facility Name
Site RU70001
City
Yaroslavl
ZIP/Postal Code
150062
Country
Russian Federation
Facility Name
Site RS38102
City
Belgrade
ZIP/Postal Code
11000
Country
Serbia
Facility Name
Site RS38105
City
Belgrade
ZIP/Postal Code
11000
Country
Serbia
Facility Name
Site RS38103
City
Belgrade
ZIP/Postal Code
11080
Country
Serbia
Facility Name
Site RS38104
City
Belgrade
Country
Serbia
Facility Name
Site RS38117
City
Krusevac
ZIP/Postal Code
37000
Country
Serbia
Facility Name
Site RS38101
City
Nis
ZIP/Postal Code
11070
Country
Serbia
Facility Name
Site SK42109
City
Kosice
ZIP/Postal Code
04001
Country
Slovakia
Facility Name
Site SK42102
City
Košice
ZIP/Postal Code
04001
Country
Slovakia
Facility Name
Site SK42113
City
Puchov
ZIP/Postal Code
02001
Country
Slovakia
Facility Name
Site SK42116
City
Senica
ZIP/Postal Code
905 01
Country
Slovakia
Facility Name
Site SI38615
City
Jesenice
ZIP/Postal Code
SI-4270
Country
Slovenia
Facility Name
Site SI38603
City
Maribor
ZIP/Postal Code
2000
Country
Slovenia
Facility Name
Site SI38619
City
Slovenj Gradec
ZIP/Postal Code
SI 2380
Country
Slovenia
Facility Name
Site SI38609
City
Šempeter pri Gorici
ZIP/Postal Code
5290
Country
Slovenia
Facility Name
Site ES34049
City
Ferrol
State/Province
A Coruna
ZIP/Postal Code
15405
Country
Spain
Facility Name
Site ES34026
City
Barcelona
ZIP/Postal Code
08907
Country
Spain
Facility Name
Site ES34039
City
Cordoba
ZIP/Postal Code
14004
Country
Spain
Facility Name
Site ES34054
City
Girona
ZIP/Postal Code
17007
Country
Spain
Facility Name
Site ES34017
City
Jaen
ZIP/Postal Code
23007
Country
Spain
Facility Name
Site ES34037
City
Madrid
ZIP/Postal Code
28046
Country
Spain
Facility Name
Site ES34010
City
Madrid
ZIP/Postal Code
28922
Country
Spain
Facility Name
Site ES34030
City
Majadahonda
ZIP/Postal Code
28222
Country
Spain
Facility Name
Site ES34041
City
Santiago de Compostela
ZIP/Postal Code
15706
Country
Spain
Facility Name
Site UA38009
City
Lviv
State/Province
Lvivska
ZIP/Postal Code
79010
Country
Ukraine
Facility Name
Site UA38021
City
Cherkasy
ZIP/Postal Code
18009
Country
Ukraine
Facility Name
Site UA38006
City
Dnipropetrovsk
ZIP/Postal Code
49005
Country
Ukraine
Facility Name
Site UA38016
City
Ivano-Frankivsk
ZIP/Postal Code
76018
Country
Ukraine
Facility Name
Site UA38011
City
Kharkiv
ZIP/Postal Code
61103
Country
Ukraine
Facility Name
Site UA38017
City
Kiev
ZIP/Postal Code
4053
Country
Ukraine
Facility Name
Site UA38007
City
Mykolaiv
Country
Ukraine
Facility Name
Site UA38008
City
Odessa
ZIP/Postal Code
6500
Country
Ukraine
Facility Name
Site UA38001
City
Ternopil
ZIP/Postal Code
46002
Country
Ukraine
Facility Name
Site UA38018
City
Uzhgorod
ZIP/Postal Code
88018
Country
Ukraine
Facility Name
Site GB44098
City
Dorchester
State/Province
Dorset
ZIP/Postal Code
DT1 2JY
Country
United Kingdom
Facility Name
Site GB44064
City
Birmingham
ZIP/Postal Code
B9 5SS
Country
United Kingdom
Facility Name
Site GB44099
City
Dartford
ZIP/Postal Code
DA2 8DA
Country
United Kingdom
Facility Name
Site GB44102
City
Kings Lynn
ZIP/Postal Code
PE30 4ET
Country
United Kingdom
Facility Name
Site GB44081
City
Leicester
ZIP/Postal Code
LE5 4PW
Country
United Kingdom
Facility Name
Site GB44086
City
London
ZIP/Postal Code
E1 1BB
Country
United Kingdom
Facility Name
Site GB44006
City
London
ZIP/Postal Code
SE5 9RS
Country
United Kingdom
Facility Name
Site GB44082
City
London
ZIP/Postal Code
SW17 0QT
Country
United Kingdom
Facility Name
Site GB44097
City
Nottingham
ZIP/Postal Code
NG5 1PB
Country
United Kingdom
Facility Name
Site GB44100
City
Orpington
ZIP/Postal Code
BR6 8ND
Country
United Kingdom
Facility Name
Site GB44101
City
Preston
ZIP/Postal Code
PR2 9HT
Country
United Kingdom
Facility Name
Site GB44080
City
Stoke-on-Trent
ZIP/Postal Code
ST4 6QG
Country
United Kingdom
Facility Name
Site GB44001
City
Swansea
ZIP/Postal Code
SA6 6NL
Country
United Kingdom

12. IPD Sharing Statement

Plan to Share IPD
Yes
IPD Sharing 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 URL
https://www.clinicalstudydatarequest.com/
Citations:
PubMed Identifier
36005278
Citation
Natale P, Palmer SC, Jaure A, Hodson EM, Ruospo M, Cooper TE, Hahn D, Saglimbene VM, Craig JC, Strippoli GF. Hypoxia-inducible factor stabilisers for the anaemia of chronic kidney disease. Cochrane Database Syst Rev. 2022 Aug 25;8(8):CD013751. doi: 10.1002/14651858.CD013751.pub2.
Results Reference
derived
Links:
URL
https://astellasclinicalstudyresults.com/hcp/study.aspx?ID=401
Description
Link to results on the Astellas Clinical Study Results website.

Learn more about this trial

Roxadustat in the Treatment of Anemia in Chronic Kidney Disease (CKD) Patients, Not on Dialysis, in Comparison to Darbepoetin Alfa

We'll reach out to this number within 24 hrs