Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT02021318
Other study ID # 1517-CL-0610
Secondary ID 2013-000951-42
Status Completed
Phase Phase 3
First received
Last updated
Start date March 12, 2014
Est. completion date November 6, 2019

Study information

Verified date April 2021
Source Astellas Pharma Inc
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial 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.


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)


Recruitment information / eligibility

Status Completed
Enrollment 616
Est. completion date November 6, 2019
Est. primary completion date March 23, 2018
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility 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

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Roxadustat
Oral tablet.
Darbepoetin alfa
Subcutaneous or intravenous injection.

Locations

Country Name City State
Austria Site AT43009 Vienna
Belarus Site BY37503 Brest
Belarus Site BY37501 Grodno
Belarus Site BY37505 Minsk
Belarus Site BY37506 Minsk
Belarus Site BY37507 Minsk
Belarus Site BY37502 Vitebsk
Bulgaria Site BG35907 Sofia
Bulgaria Site BG35927 Sofia
Bulgaria Site BG35916 Varna
Croatia Site HR38505 Karlovac
Croatia Site HR38504 Slavonski Brod
Croatia Site HR38502 Zagreb
Croatia Site HR38509 Zagreb
Croatia Site HR38510 Zagreb
Czechia Site CZ42008 Liberec
Czechia Site CZ42018 Novy Jicin CZ
Czechia Site CZ42021 Prague
Finland Site FI35810 Helsinki
France Site FR33004 Avignon
France Site FR33009 Colmar
France Site FR33010 Grenoble
France Site FR33062 Limoges
France Site FR33012 Lyon
France Site FR33007 Saint-Priest-en-Jarez
Georgia Site GE99502 Tbilisi
Georgia Site GE99503 Tbilisi
Georgia Site GE99504 Tbilisi
Germany Site DE49073 Cloppenburg
Germany Site DE49054 Düsseldorf
Germany Site DE49065 Hamburg
Germany Site DE49075 Heilbronn
Germany Site DE49057 Hoyerswerda
Hungary Site HU36028 Budapest
Hungary Site HU36029 Budapest
Hungary Site HU36027 Kistarcsa
Hungary Site HU36008 Pecs
Hungary Site HU36046 Velence
Ireland Site IE35301 Cork
Israel Site IL97202 Be'er Ya'akov
Israel Site IL97215 Haifa
Latvia Site LV37101 Riga
Latvia Site LV37104 Ventspils
Montenegro Site ME38202 Niksic
Montenegro Site ME38201 Podgorica
Netherlands Site NL31005 Utrecht
North Macedonia Site MK38901 Skopje
North Macedonia Site MK38903 Struga
Poland Site PL48001 Krakow
Poland Site PL48066 Pulawy
Poland Site PL48013 Szczecin
Poland Site PL48007 Tarnow
Poland Site PL48004 Warszawa
Poland Site PL48009 Wroclaw
Poland Site PL48059 Zamosc
Portugal Site PT35120 Almada
Portugal Site PT35131 Braga
Portugal Site PT35112 Carnaxide
Portugal Site PT35119 Evora
Portugal Site PT35118 Lisboa
Portugal Site PT35133 Matosinhos
Portugal Site PT35122 Setubal
Portugal Site PT35132 Vila Nova de Gaia
Romania Site RO40003 Bucharest
Romania Site RO40021 Bucharest
Romania Site RO40012 Bucuresti
Romania Site RO40004 Oradea
Russian Federation Site RU70024 Chelyabinsk
Russian Federation Site RU70054 Irkutsk
Russian Federation Site RU70006 Moscow
Russian Federation Site RU70047 Moscow
Russian Federation Site RU70003 Nizhny Novgorod
Russian Federation Site RU70004 Omsk
Russian Federation Site RU70014 Rostov-on-Don
Russian Federation Site RU70002 Saint Petersburg
Russian Federation Site RU70011 Saint Petersburg
Russian Federation Site RU70060 Saratov
Russian Federation Site RU70001 Yaroslavl
Russian Federation Site RU70057 Yaroslavl
Serbia Site RS38102 Belgrade
Serbia Site RS38103 Belgrade
Serbia Site RS38104 Belgrade
Serbia Site RS38105 Belgrade
Serbia Site RS38117 Krusevac
Serbia Site RS38101 Nis
Slovakia Site SK42102 Košice
Slovakia Site SK42109 Kosice
Slovakia Site SK42113 Puchov
Slovakia Site SK42116 Senica
Slovenia Site SI38609 Šempeter pri Gorici
Slovenia Site SI38615 Jesenice
Slovenia Site SI38603 Maribor
Slovenia Site SI38619 Slovenj Gradec
Spain Site ES34026 Barcelona
Spain Site ES34039 Cordoba
Spain Site ES34049 Ferrol A Coruna
Spain Site ES34054 Girona
Spain Site ES34017 Jaen
Spain Site ES34010 Madrid
Spain Site ES34037 Madrid
Spain Site ES34030 Majadahonda
Spain Site ES34041 Santiago de Compostela
Ukraine Site UA38021 Cherkasy
Ukraine Site UA38006 Dnipropetrovsk
Ukraine Site UA38016 Ivano-Frankivsk
Ukraine Site UA38011 Kharkiv
Ukraine Site UA38017 Kiev
Ukraine Site UA38009 Lviv Lvivska
Ukraine Site UA38007 Mykolaiv
Ukraine Site UA38008 Odessa
Ukraine Site UA38001 Ternopil
Ukraine Site UA38018 Uzhgorod
United Kingdom Site GB44064 Birmingham
United Kingdom Site GB44099 Dartford
United Kingdom Site GB44098 Dorchester Dorset
United Kingdom Site GB44102 Kings Lynn
United Kingdom Site GB44081 Leicester
United Kingdom Site GB44006 London
United Kingdom Site GB44082 London
United Kingdom Site GB44086 London
United Kingdom Site GB44097 Nottingham
United Kingdom Site GB44100 Orpington
United Kingdom Site GB44101 Preston
United Kingdom Site GB44080 Stoke-on-Trent
United Kingdom Site GB44001 Swansea

Sponsors (2)

Lead Sponsor Collaborator
Astellas Pharma Europe B.V. FibroGen

Countries where clinical trial is conducted

Austria,  Belarus,  Bulgaria,  Croatia,  Czechia,  Finland,  France,  Georgia,  Germany,  Hungary,  Ireland,  Israel,  Latvia,  Montenegro,  Netherlands,  North Macedonia,  Poland,  Portugal,  Romania,  Russian Federation,  Serbia,  Slovakia,  Slovenia,  Spain,  Ukraine,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary 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). Baseline to week 24
Secondary 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). Baseline and weeks 28 to 36
Secondary 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. Baseline and weeks 12 to 28
Secondary 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. Weeks 6, 12, 18, 24, 30 and 36
Secondary 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. Baseline and weeks 12 to 28
Secondary 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. Baseline and weeks 12 to 28
Secondary 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). Baseline and weeks 20 to 28
Secondary 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. Weeks 1 to 36
Secondary 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. Baseline and weeks 20 to 28
Secondary 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. Weeks 1 to 36
Secondary 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). Baseline and weeks 28 to 52
Secondary 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. Weeks 1 to 24
Secondary 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. Weeks 1 to 24
Secondary 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). Weeks 28 to 36, 44 to 52, 72 to 80 and 96 to 104
Secondary 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). 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
Secondary 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). Baseline and weeks 28 to 36, 44 to 52, 72 to 80 and 96 to 104
Secondary 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. Weeks 28 to 36, 44 to 52 and 96 to 104
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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). Baseline to EOT (up to week 104)
Secondary 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). Baseline to EOT (up to week 104)
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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. Baseline to EOT (up to week 104)
Secondary 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. Baseline to EOT (up to week 104)
Secondary 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). Baseline to EOT (up to week 104)
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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. Baseline to EOT (up to week 104)
Secondary 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. Weeks 37 to 52 and 53 to 104
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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). Day 1 to week 36, weeks 37 to 52, weeks 53 to 104, efficacy emergent period (up to week 104)
Secondary 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. Baseline and weeks 8, 28, 52, 104
Secondary 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. Baseline and weeks 8, 28, 52, 104
Secondary 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. Baseline and weeks 8, 28, 52, 104
Secondary 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. Baseline and weeks 8, 28, 52, 104
Secondary 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. Baseline and weeks 8, 28, 52, 104
Secondary 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. Baseline and weeks 8, 28, 52, 104
Secondary 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. Weeks 12 to 28 and 36 to 52
Secondary 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. Weeks 12 to 28 and 36 to 52
Secondary 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. Baseline, weeks 12 to 28 and 36 to 52
Secondary 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. Baseline, weeks 12 to 28 and 36 to 52
Secondary 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. Baseline, weeks 12 to 28 and 36 to 52
Secondary 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. Baseline, weeks 12 to 28 and 36 to 52
Secondary 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. Baseline and weeks 12 to 28
Secondary 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). Baseline, weeks 12 to 28 and 36 to 52
Secondary 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. Baseline, weeks 12 to 28 and 36 to 52
Secondary 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)]. Baseline, weeks 12 to 28 and 36 to 52
Secondary 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. Baseline, weeks 12 to 28 and 36 to 52
Secondary 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. Weeks 8, 12, 28, 52, 76, 104, last assessment (week 108)
Secondary 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. 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)
Secondary 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. Baseline and weeks 4, 8, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 92, 100, 104 and EOS (up to 108 weeks)
Secondary 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. Baseline and weeks 12, 28, 36, 44, 60, 84, 104 and EOS (up to 108 weeks)
Secondary 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. 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)
Secondary 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. 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)
Secondary 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. Baseline up to EOS (up to week 108)
Secondary 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. Baseline and weeks 12, 24, 36, 52, 64, 76, 88 and 104
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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. Baseline up to EOS (up to week 108)
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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. Year 0.5, 1, 1.5 and 2
Secondary 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. From first dose of study drug up to end of study (up to week 108)
See also
  Status Clinical Trial Phase
Completed NCT01887600 - Roxadustat in the Treatment of Anemia in Chronic Kidney Disease Patients Not Requiring Dialysis Phase 3