A Study of AGS-16C3F vs. Axitinib in Metastatic Renal Cell Carcinoma

Sponsor
Astellas Pharma Global Development, Inc. (Industry)
Overall Status
Completed
CT.gov ID
NCT02639182
Collaborator
(none)
133
26
2
53
5.1
0.1

Study Details

Study Description

Brief Summary

The purpose of this study was to evaluate the progression free survival (PFS), based on investigator radiologic review, of AGS-16C3F compared to axitinib in subjects with metastatic renal cell carcinoma.

Condition or Disease Intervention/Treatment Phase
Phase 2

Study Design

Study Type:
Interventional
Actual Enrollment :
133 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
A Multi-Center, Open Label, Randomized Phase 2 Study of AGS-16C3F vs. Axitinib in Metastatic Renal Cell Carcinoma
Actual Study Start Date :
May 3, 2016
Actual Primary Completion Date :
Oct 2, 2020
Actual Study Completion Date :
Oct 2, 2020

Arms and Interventions

Arm Intervention/Treatment
Experimental: AGS-16C3F

Participants received 1.8 milligram per kilogram (mg/kg) of AGS-16C3F once every three weeks by single intravenous (IV) infusion.

Drug: AGS-16C3F
Intravenous (IV) infusion

Active Comparator: Axitinib

Participants received 2 to 10 milligram (mg) of axitinib twice daily by oral administration as defined in the product label and per local institutional guidelines.

Drug: Axitinib
Oral
Other Names:
  • Inlyta®
  • Outcome Measures

    Primary Outcome Measures

    1. Progression Free Survival (PFS) as Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Investigator Radiologic Review [From date of randomization to the earliest of either documented disease progression or death from any cause (up to 53 months)]

      PFS was defined as the time from the date of randomization to the earliest of documented disease progression as defined by RECIST v.1.1 or death from any cause. PFS was analysed using Kaplan-Meier estimates. Progressive disease (PD) was defined as at least a 20% increase in the sum of diameters (longest for non-nodal lesions, short axis for nodal lesions) of the target lesions taking as reference the smallest sum on study. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 millimeter (mm). The appearance of 1 or more new lesions is also considered progression. Participants were censored if there were 2 or more than 2 consecutive missing assesments immediately prior to death or progression; or no death and no postebaseline assesments; or if ininitiated non protocol anticancer treatment prior to death or prior to documentation of disease progression; or alive and not progressed.

    Secondary Outcome Measures

    1. PFS Per RECIST v1.1 as Assessed by Blinded Central Radiology Assessment [From date of randomization to the earliest of either documented disease progression or death from any cause (up to 40 months)]

      PFS was defined as the time from the date of randomization to the earliest of documented disease progression as defined by RECIST v.1.1 or death from any cause. PFS was analysed using Kaplan-Meier estimates. PD was defined as at least a 20% increase in the sum of diameters (longest for non-nodal lesions, short axis for nodal lesions) of the target lesions taking as reference the smallest sum on study. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5mm. The appearance of 1 or more new lesions is also considered progression. Participants were censored if there were 2 or more than 2 consecutive missing assessments immediately prior to death or progression; or no death and no postebaseline assessments; or if initiated non protocol anticancer treatment prior to death or prior to documentation of disease progression; or alive and not progressed.

    2. Objective Response Rate (ORR) Based on the Investigator's Radiographic Assessment [From date of randomization until data cutoff date of 21 August 2019 (up to 40 months)]

      ORR was defined as the percentage of participants who had a best overall response of complete response (CR) or partial response (PR). CR was defined as disappearance of all target and nontarget lesions. Any pathological lymph nodes (whether target or nontarget) with reduction in short axis to <10mm from baseline measurement. PR was defined as at least a 30% decrease in the sum of diameters (longest for nonnodal lesions, short axis for nodal lesions) of target lesions taking as reference to the baseline sum of diameters.

    3. Duration of Response (DOR) Based on the Investigator's Radiographic Assessment [From date of first objective response until data cutoff date of 21 August 2019 (up to 40 months)]

      DOR was defined as the time from the date of the first response of CR or PR (whichever was first recorded) to the first date of documented PD or death due to any cause. DOR was analysed using Kaplan-Meier estimates. CR and PR were defined in outcome measure 3 and PD was defined in outcome measures 1 and 2. Participants were censored if there were 2 or more than 2 consecutive missing assesments immediately prior to death or progression; or no death and no postebaseline assesments; or if ininitiated non protocol anticancer treatment prior to death or prior to documentation of disease progression; or alive and not progressed.

    4. Overall Survival (OS) [Date of randomization until the date of death from any cause (up to 53 months)]

      OS was defined as the time from the date of randomization until the date of death from any cause. OS was analysed using Kaplan-Meier estimates. Participants were censored if there was no death and no postbaseline contact or no death and at least one postbaseline follow-up.

    5. Disease Control Rate (DCR) Based on the Investigator's Radiographic Assessment [Date of randomization until data cutoff date of 21 August 2019 (up to 40 months)]

      DCR was defined as the percentage of patients who had a best overall response of CR, PR or at least 6 months with stable disease (SD). CR was defined as disappearance of all target and nontarget lesions. Any pathological lymph nodes (whether target or nontarget) with reduction in short axis to <10mm from baseline measurement. PR was defined as at least a 30% decrease in the sum of diameters (longest for nonnodal lesions, short axis for nodal lesions) of target lesions taking as reference to the baseline sum of diameters. SD was defined as neither sufficient decrease to qualify for PR nor sufficient increase to qualify for progressive disease taking as reference the smallest sum of diameters while on study drug.

    6. Number of Participants With Adverse Events [From first dose up to 53 months]

      AE was defined as any untoward medical occurrence in a participant administered a study drug or has undergone study procedures and which did not necessarily have a causal relationship with this treatment. An abnormality identified during a medical test (e.g., laboratory parameter, vital sign, ECG data, and physical exam) was defined as an AE only if the abnormality induces clinical signs or symptoms or requires active intervention or requires interruption or discontinuation of study medication or the abnormality or investigational value is clinically significant in the opinion of the investigator. AE was considered "serious" if it results in death or is life threatening results in persistent or significant disability/incapacity or substantial disruption of the ability to conduct normal life functions or results in congenital anomaly, or birth defect requires inpatient hospitalization or leads to prolongation of hospitalization or other medically important events.

    7. Maximum Observed Serum Concentration (Cmax) of Antibody Drug Conjugate (ADC) [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      Cmax of ADC was reported.

    8. Mean Predose Serum Concentration (Ctrough) of ADC [Predose on cycle 2, 3, 4, 6, 14, and 18 (each cycle is 21 days)]

      Ctrough of ADC was reported.

    9. Time to Maximum Observed Serum Concentration (Tmax) of ADC [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      Tmax of ADC was reported.

    10. Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of ADC [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      AUC (0 to 21) of ADC was reported.

    11. Terminal Elimination Half-life (t1/2) of ADC [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      t1/2 of ADC was reported.

    12. Maximum Serum Concentration (Cmax) of Total Antibody (TAb) [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      Cmax of TAb was reported.

    13. Mean Predose Serum Concnetration (Ctrough) of TAb [Predose on cycle 2, 3, 4, 6, 14, and 18 (each cycle is 21 days)]

      Ctrough of TAb was reported.

    14. Time to Maximum Observed Serum Concentration (Tmax) of Tab [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      Tmax of TAb was reported.

    15. Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of TAb [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      AUC (0 to 21) of TAb was reporetd.

    16. Terminal Elimination Half-life (t1/2) of Tab [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      t1/2 of TAb was reported.

    17. Maximum Serum Concentration (Cmax) of Cysteine Adduct of Maleimidocaproyl Monomethyl Auristatin F (Cys-mcMMAF) [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      Cmax of Cys-mcMMAF was reported.

    18. Mean Predose Serum Concnetration (Ctrough) of Cys-mcMMAF [Predose on cycle 2, 3, 4, 6, 14, and 18 (each cycle is 21 days)]

      Ctrough of Cys-mcMMAF was reported.

    19. Time to Maximum Observed Serum Concentration (Tmax) of Cys-mcMMAF [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      Tmax of Cys-mcMMAF was reported.

    20. Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of Cys-mcMMAF [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      AUC (0 to 21) of Cys-mcMMAF was reporetd.

    21. Terminal Elimination Half-life (t1/2) of Cys-mcMMAF [Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)]

      t1/2 of Cys-mcMMAF was reported

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    18 Years and Older
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    • Histologically confirmed diagnosis of RCC

    • Non-clear subjects must be ENPP3 positive, defined as IHC H-score ≥15

    • Has evidence of progression on or after the last regimen received:

    • Clear cell subject: must have received at least 2 prior systemic regimens, one of which is an anti-VEGF agent.

    • Non-clear cell subject: must have received at least one prior anti-VEGF regimen

    • Has measurable disease according to Response Criteria for Solid Tumors (RECIST v.1.1)

    • Has Eastern Cooperative Group (ECOG) performance status of 0 or 1

    • Has archive tumor tissue from primary tumor or metastatic site (excluding bone), for which the source and availability have been confirmed.

    • If no archive tissue is available, the subject may elect to have a biopsy performed to obtain tissue.

    • Has adequate organ function including:

    • Hematopoietic function as follows:

    1. Absolute neutrophil count (ANC) ≥ 1.5 x 10 9/L

    2. Platelet count ≥ 100 x 10 9/L

    3. Hemoglobin ≥ 9 g/dL (transfusions are allowed)

    • Renal Function as follows:
    1. Creatinine ≤ 1.5 x upper limit of normal (ULN), or calculated glomerular filtration rate (GFR) > 40 mL/min (Cockcroft-Gault) if creatinine > 1.5x ULN
    • Hepatic function, as follows:
    1. Aspartate aminotransferase (AST) and Alanine aminotransferase (ALT) ≤ 2.5 x ULN or ≤ 5x ULN if known liver metastases

    2. Total bilirubin ≤ 1.5 x ULN

    • Prothrombin time (PT) and activated partial thromboplastin time (aPTT) levels ≤1.5 x ULN. If institution does not report PT value, the international normalization ratio (INR) must be ≤ ULN.

    • If subject is receiving Coumadin (warfarin), a stable international normalization ratio (INR) of 2-3 is required.

    • No clinical symptoms of hypothyroidism

    • Urine Protein to Creatinine Ratio (uPCR) < 2.0

    • If uPCR ≥ 2.0 then a 24-hour urine collection can be performed to qualify. If this is performed to qualify, the protein result must be < 2 g per 24 hours.

    • Female subject must either:

    • Be of non-childbearing potential:

    1. post-menopausal (defined as at least 1 year without any menses) prior to Screening, or

    2. documented surgically sterile

    • Or, if of childbearing potential,
    1. Agree not to try to become pregnant during the study and for 6 months after the final study drug administration

    2. And have a negative serum pregnancy test ≤ 10 days of cycle 1, day 1 (C1D1)

    • And, if heterosexually active, agree to consistently use 2 forms of highly effective birth control* (at least one of which must be a barrier method) starting at Screening and throughout the study period and for 6 months after the final study drug administration.

    • Female subject must agree not to breastfeed starting at Screening and throughout the study period, and for 6 months after the final study drug administration.

    • Female subject must not donate ova starting at Screening and throughout the study period, and for 6 months after the final study drug administration.

    • Male subject and their female spouse/partners who are of childbearing potential must be using highly effective contraception* consisting of 2 forms of birth control (at least one of which must be a barrier method) starting at Screening and continue throughout the study period, and for 6 months after the final study drug administration

    • Male subject must not donate sperm starting at Screening and throughout the study period and, for 6 months after the final study drug administration

    Note: *Highly effective forms of birth control include:
    • Consistent and correct usage of established oral contraception.

    • Established intrauterine device (IUD) or intrauterine system (IUS).

    • Barrier methods of contraception: condom or occlusive cap (diaphragm or cervical/vault caps) with spermicidal foam/gel/film/cream/suppository

    Exclusion Criteria:
    • Has previously been treated with axitinib, AGS-16C3F, or AGS-16M8F

    • Has untreated brain metastasis. In the case of a solitary brain metastasis which has been resected, there must be evidence of a disease-free interval of at least 3 months post-surgery. For brain metastases treated with whole brain or stereotactic radiation therapy, brain imaging must be stable > 3 months. All subjects previously treated for brain metastases must be stable off corticosteroid therapy for at least 28 days prior to C1D1.

    • Has uncontrolled hypertension defined as blood pressure > 150/90 on medication(s) by 2 blood pressure readings taken at least 1 hour apart.

    • Has gastrointestinal abnormalities including:

    • inability to take oral medication;

    • requirement for intravenous alimentation;

    • prior surgical procedures affecting absorption including total gastric resection;

    • active gastrointestinal bleeding, unrelated to cancer, as evidenced by hematemesis, hematochezia or melena in the past 3 months without evidence of resolution documented by endoscopy or colonoscopy;

    • malabsorption syndromes such as celiac disease, cystic fibrosis, inflammatory bowel disease, systemic sclerosis, and carcinoid syndrome

    • Has ocular conditions such as:

    • Active infection or corneal ulcer

    • Monocularity

    • Visual acuity of 20/70 or worse in both eyes

    • History of corneal transplantation

    • Contact lens dependent (if using contact lens, must be able to switch to glasses during the entire study duration)

    • Uncontrolled glaucoma (topical medications allowed)

    • Uncontrolled or active ocular problems (e.g., retinopathy, macular edema, active uveitis, wet macular degeneration) requiring surgery, laser treatment, or intravitreal injections

    • Papilledema or other active optic nerve disorder

    • Has used any investigational drug (including marketed drugs not approved for this indication) ≤ 14 days of C1D1. No time limit applies to the use of marketed drugs approved for this indication provided that the subject has progressed on the treatment and all toxicities attributable to the drug have resolved, returned to baseline or stabilized.

    • Has known sensitivity to any of the ingredients of:

    • investigational product AGS-16C3F and/or,

    • Inlyta® (axitinib) and/or,

    • 1% prednisolone acetate ophthalmic suspension and any other corticosteroids.

    • Is currently using (i.e., within 14-days prior to first dose) drugs that are known strong CYP3A4/5 inhibitors / inducers.

    • Thromboembolic event (e.g., deep vein thrombosis [DVT] and pulmonary embolism [PE]) ≤ 4 weeks of C1D1.

    • Subjects who had a thromboembolic event ≤ 4 weeks of C1D1 must be receiving adequate anticoagulation treatment for at least 2 weeks before C1D1 and must continue as clinically indicated post first dose

    • Has history bleeding disorders (e.g., pulmonary hemorrhage, significant hemoptysis, menometrorrhagia not responding to hormonal treatment) ≤ 2 months before C1D1

    • Has active angina or Class III or IV Congestive Heart Failure (New York Heart Association CHF Functional Classification System) or clinically significant cardiac disease within 6 months of randomization, including myocardial infarction, unstable angina, Grade 2 or greater peripheral vascular disease, congestive heart failure, or arrhythmias not controlled by medication.

    • Had major surgery ≤ 4 weeks of C1D1

    • Is pregnant (confirmed by positive serum pregnancy test) or lactating

    • Has active infection requiring treatment with systemic (intravenous or oral) anti-infectives (antibiotic, antifungal, or antiviral agent) ≤ 10 days of C1D1

    • Is unwilling or unable to comply with study requirements

    • Has any medical or psychiatric disorder that compromises the ability of the subject to give written informed consent, and/or comply with the study procedures.

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Site US01026 Tucson Arizona United States 85719
    2 Site US01008 La Jolla California United States 92093
    3 Site US01007 Los Angeles California United States 90033
    4 Site US01020 Los Angeles California United States 90095
    5 Site US01019 Palo Alto California United States 94305
    6 Site US01010 Atlanta Georgia United States 30322
    7 Site US01023 Baltimore Maryland United States 21201
    8 Site US01002 Boston Massachusetts United States 02215
    9 Site US01004 Ann Arbor Michigan United States 48109
    10 Site US01013 Detroit Michigan United States 48202
    11 Site US01012 Omaha Nebraska United States 68130
    12 Site US01006 Buffalo New York United States 14263
    13 Site US01022 Durham North Carolina United States 27710
    14 Site US01017 Portland Oregon United States 97213
    15 Site US01021 Pittsburgh Pennsylvania United States 15232
    16 Site US01011 Charleston South Carolina United States 29425
    17 Site US01003 Houston Texas United States 77030
    18 Site US01014 Temple Texas United States 76508
    19 Site US01001 Seattle Washington United States 98109
    20 Site US01009 Milwaukee Wisconsin United States 53226
    21 Site CA02006 Calgary Alberta Canada T2N 4N2
    22 Site CA02004 Edmonton Alberta Canada T6G 1Z2
    23 Site CA02005 Kelowna British Columbia Canada V1Y 5L3
    24 Site CA02001 Vancouver British Columbia Canada V5Z 4E6
    25 Site CA02002 Hamilton Ontario Canada L8V 5C2
    26 Site CA02008 London Ontario Canada N6A 5W9

    Sponsors and Collaborators

    • Astellas Pharma Global Development, Inc.

    Investigators

    • Study Director: Associate Medical Director, Astellas Pharma Global Development, Inc.

    Study Documents (Full-Text)

    More Information

    Additional Information:

    Publications

    None provided.
    Responsible Party:
    Astellas Pharma Global Development, Inc.
    ClinicalTrials.gov Identifier:
    NCT02639182
    Other Study ID Numbers:
    • AGS-16C3F-15-3
    First Posted:
    Dec 24, 2015
    Last Update Posted:
    Nov 10, 2021
    Last Verified:
    Nov 1, 2021
    Individual Participant Data (IPD) Sharing Statement:
    Yes
    Plan to Share IPD:
    Yes
    Studies a U.S. FDA-regulated Drug Product:
    Yes
    Studies a U.S. FDA-regulated Device Product:
    No
    Keywords provided by Astellas Pharma Global Development, Inc.
    Additional relevant MeSH terms:

    Study Results

    Participant Flow

    Recruitment Details Participants who were atleast 18 years of age with all histologies of confirmed renal cell carcinoma and evidence of progression on or after the last regimen received were enrolled.
    Pre-assignment Detail Randomization was stratified by Eastern Cooperative Oncology Group (ECOG) performance status (0 or 1), the number of prior systemic RCC regimens (2 or > 2) and Renal cell carcinoma (RCC), histology (clear or nonclear cell).
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 milligram per kilogram (mg/kg) of AGS-16C3F once every three weeks by single (60-minute) intravenous (IV) infusion. Participants received axitinib at a starting dose of 5 milligram (mg) orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Period Title: Overall Study
    STARTED 67 66
    Treated 66 65
    COMPLETED 0 0
    NOT COMPLETED 67 66

    Baseline Characteristics

    Arm/Group Title AGS-16C3F Axitinib Total
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines. Total of all reporting groups
    Overall Participants 67 66 133
    Age (Years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [Years]
    62.3
    (9.1)
    61.1
    (8.9)
    61.7
    (9)
    Sex: Female, Male (Count of Participants)
    Female
    18
    26.9%
    17
    25.8%
    35
    26.3%
    Male
    49
    73.1%
    49
    74.2%
    98
    73.7%
    Ethnicity (NIH/OMB) (Count of Participants)
    Hispanic or Latino
    3
    4.5%
    3
    4.5%
    6
    4.5%
    Not Hispanic or Latino
    64
    95.5%
    62
    93.9%
    126
    94.7%
    Unknown or Not Reported
    0
    0%
    1
    1.5%
    1
    0.8%
    Race/Ethnicity, Customized (Count of Participants)
    White
    59
    88.1%
    56
    84.8%
    115
    86.5%
    Asian
    6
    9%
    7
    10.6%
    13
    9.8%
    American Indian or Alaska Native
    1
    1.5%
    1
    1.5%
    2
    1.5%
    Other
    1
    1.5%
    2
    3%
    3
    2.3%
    Eastern Cooperative Oncology Group (ECOG) Performance Status (Count of Participants)
    0
    19
    28.4%
    19
    28.8%
    38
    28.6%
    1
    48
    71.6%
    47
    71.2%
    95
    71.4%
    Histological Type (Count of Participants)
    Clear cell
    55
    82.1%
    55
    83.3%
    110
    82.7%
    Non-clear cell
    12
    17.9%
    11
    16.7%
    23
    17.3%
    Prognostic Risk Group (Count of Participants)
    Favorable (0 risk factors)
    5
    7.5%
    10
    15.2%
    15
    11.3%
    Intermediate (1-2 risk factors)
    48
    71.6%
    43
    65.2%
    91
    68.4%
    Poor (>=3 risk factors)
    14
    20.9%
    13
    19.7%
    27
    20.3%
    Number of Prior Systemic Renal cell carcinoma (RCC) Treatment Regimens (Count of Participants)
    2 if clear cell or 1 if non-clear cell histology
    35
    52.2%
    33
    50%
    68
    51.1%
    >2 if clear cell or >1 if non-clear cell histology
    32
    47.8%
    33
    50%
    65
    48.9%

    Outcome Measures

    1. Primary Outcome
    Title Progression Free Survival (PFS) as Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Investigator Radiologic Review
    Description PFS was defined as the time from the date of randomization to the earliest of documented disease progression as defined by RECIST v.1.1 or death from any cause. PFS was analysed using Kaplan-Meier estimates. Progressive disease (PD) was defined as at least a 20% increase in the sum of diameters (longest for non-nodal lesions, short axis for nodal lesions) of the target lesions taking as reference the smallest sum on study. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 millimeter (mm). The appearance of 1 or more new lesions is also considered progression. Participants were censored if there were 2 or more than 2 consecutive missing assesments immediately prior to death or progression; or no death and no postebaseline assesments; or if ininitiated non protocol anticancer treatment prior to death or prior to documentation of disease progression; or alive and not progressed.
    Time Frame From date of randomization to the earliest of either documented disease progression or death from any cause (up to 53 months)

    Outcome Measure Data

    Analysis Population Description
    FAS Population
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Measure Participants 67 66
    Median (95% Confidence Interval) [Months]
    2.9
    5.7
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection AGS-16C3F, Axitinib
    Comments
    Type of Statistical Test Superiority
    Comments The difference between treatment groups was tested using a stratified log rank test at a one-sided 0.1 significance level.
    Statistical Test of Hypothesis p-Value 0.983
    Comments The stratification factors were the ECOG PS at baseline and the number of prior systemic renal cell carcinoma (RCC) regimens, without any other covariate(s). The model contained terms for treatment group and stratum.
    Method Log Rank
    Comments
    2. Secondary Outcome
    Title PFS Per RECIST v1.1 as Assessed by Blinded Central Radiology Assessment
    Description PFS was defined as the time from the date of randomization to the earliest of documented disease progression as defined by RECIST v.1.1 or death from any cause. PFS was analysed using Kaplan-Meier estimates. PD was defined as at least a 20% increase in the sum of diameters (longest for non-nodal lesions, short axis for nodal lesions) of the target lesions taking as reference the smallest sum on study. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5mm. The appearance of 1 or more new lesions is also considered progression. Participants were censored if there were 2 or more than 2 consecutive missing assessments immediately prior to death or progression; or no death and no postebaseline assessments; or if initiated non protocol anticancer treatment prior to death or prior to documentation of disease progression; or alive and not progressed.
    Time Frame From date of randomization to the earliest of either documented disease progression or death from any cause (up to 40 months)

    Outcome Measure Data

    Analysis Population Description
    FAS Population
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Measure Participants 67 66
    Median (95% Confidence Interval) [Months]
    3.5
    4.5
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection AGS-16C3F, Axitinib
    Comments
    Type of Statistical Test Superiority
    Comments The difference between treatment groups was tested using a stratified log rank test at a one-sided 0.1 significance level.
    Statistical Test of Hypothesis p-Value 0.110
    Comments The stratification factors were the ECOG PS at baseline and the number of prior systemic RCC regimens, without any other covariate(s). The model contained terms for treatment group and stratum.
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 1.423
    Confidence Interval (2-Sided) 95%
    0.924 to 2.192
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    3. Secondary Outcome
    Title Objective Response Rate (ORR) Based on the Investigator's Radiographic Assessment
    Description ORR was defined as the percentage of participants who had a best overall response of complete response (CR) or partial response (PR). CR was defined as disappearance of all target and nontarget lesions. Any pathological lymph nodes (whether target or nontarget) with reduction in short axis to <10mm from baseline measurement. PR was defined as at least a 30% decrease in the sum of diameters (longest for nonnodal lesions, short axis for nodal lesions) of target lesions taking as reference to the baseline sum of diameters.
    Time Frame From date of randomization until data cutoff date of 21 August 2019 (up to 40 months)

    Outcome Measure Data

    Analysis Population Description
    FAS Population
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Measure Participants 67 66
    Number (95% Confidence Interval) [Percentage of Participants]
    7.5
    11.2%
    18.2
    27.6%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection AGS-16C3F, Axitinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.062
    Comments A Cochran-Mantel-Haenszel (CMH) analysis of the ORR, stratified for baseline ECOG-PS and number of prior systemic therapies for RCC, was conducted at a two-sided significance level of 0.05.
    Method Cochran-Mantel-Haenszel
    Comments Stratified Mantel-Haenszel estimate of the odds ratio for experiencing objective response, with the axitinib arm as the reference level, was reported.
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 0.4
    Confidence Interval (2-Sided) 95%
    0.1 to 1.1
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    4. Secondary Outcome
    Title Duration of Response (DOR) Based on the Investigator's Radiographic Assessment
    Description DOR was defined as the time from the date of the first response of CR or PR (whichever was first recorded) to the first date of documented PD or death due to any cause. DOR was analysed using Kaplan-Meier estimates. CR and PR were defined in outcome measure 3 and PD was defined in outcome measures 1 and 2. Participants were censored if there were 2 or more than 2 consecutive missing assesments immediately prior to death or progression; or no death and no postebaseline assesments; or if ininitiated non protocol anticancer treatment prior to death or prior to documentation of disease progression; or alive and not progressed.
    Time Frame From date of first objective response until data cutoff date of 21 August 2019 (up to 40 months)

    Outcome Measure Data

    Analysis Population Description
    FAS Population
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Measure Participants 67 66
    Median (95% Confidence Interval) [Months]
    6.8
    6.7
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection AGS-16C3F, Axitinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.439
    Comments The stratification factors were the ECOG PS at baseline and the number of prior systemic RCC regimens, without any other covariate(s). The model contained terms for treatment group and stratum.
    Method Log Rank
    Comments The difference between treatment groups was tested using a stratified log rank test at a one-sided 0.1 significance level.
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.376
    Confidence Interval () 95%
    0.031 to 4.482
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    5. Secondary Outcome
    Title Overall Survival (OS)
    Description OS was defined as the time from the date of randomization until the date of death from any cause. OS was analysed using Kaplan-Meier estimates. Participants were censored if there was no death and no postbaseline contact or no death and at least one postbaseline follow-up.
    Time Frame Date of randomization until the date of death from any cause (up to 53 months)

    Outcome Measure Data

    Analysis Population Description
    FAS Population
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Measure Participants 67 66
    Median (95% Confidence Interval) [Months]
    13.1
    15.5
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection AGS-16C3F, Axitinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.746
    Comments Test conducted at a 2-sided significance level of 0.05. The stratification factors were the ECOG PS at baseline and number of prior systemic RCC regimens.
    Method Log Rank
    Comments
    6. Secondary Outcome
    Title Disease Control Rate (DCR) Based on the Investigator's Radiographic Assessment
    Description DCR was defined as the percentage of patients who had a best overall response of CR, PR or at least 6 months with stable disease (SD). CR was defined as disappearance of all target and nontarget lesions. Any pathological lymph nodes (whether target or nontarget) with reduction in short axis to <10mm from baseline measurement. PR was defined as at least a 30% decrease in the sum of diameters (longest for nonnodal lesions, short axis for nodal lesions) of target lesions taking as reference to the baseline sum of diameters. SD was defined as neither sufficient decrease to qualify for PR nor sufficient increase to qualify for progressive disease taking as reference the smallest sum of diameters while on study drug.
    Time Frame Date of randomization until data cutoff date of 21 August 2019 (up to 40 months)

    Outcome Measure Data

    Analysis Population Description
    FAS Population
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Measure Participants 67 66
    Number (95% Confidence Interval) [Percentage of Participants]
    13.4
    20%
    22.7
    34.4%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection AGS-16C3F, Axitinib
    Comments
    Type of Statistical Test Superiority
    Comments A CMH analysis of the DCR, stratified for baseline ECOG-PS and number of prior systemic therapies for RCC, was conducted at a two-sided significance level of 0.05.
    Statistical Test of Hypothesis p-Value 0.152
    Comments The stratified Mantel-Haenszel estimate of the odds ratio for experiencing objective response, with the axitinib arm as the reference level, was reported as a measure of relative treatment effect, along with its two-sided 95% CI.
    Method Cochran-Mantel-Haenszel
    Comments
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 0.5
    Confidence Interval (2-Sided) 95%
    0.2 to 1.3
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    7. Secondary Outcome
    Title Number of Participants With Adverse Events
    Description AE was defined as any untoward medical occurrence in a participant administered a study drug or has undergone study procedures and which did not necessarily have a causal relationship with this treatment. An abnormality identified during a medical test (e.g., laboratory parameter, vital sign, ECG data, and physical exam) was defined as an AE only if the abnormality induces clinical signs or symptoms or requires active intervention or requires interruption or discontinuation of study medication or the abnormality or investigational value is clinically significant in the opinion of the investigator. AE was considered "serious" if it results in death or is life threatening results in persistent or significant disability/incapacity or substantial disruption of the ability to conduct normal life functions or results in congenital anomaly, or birth defect requires inpatient hospitalization or leads to prolongation of hospitalization or other medically important events.
    Time Frame From first dose up to 53 months

    Outcome Measure Data

    Analysis Population Description
    Safety Population: All randomized participants who received at least one dose of study drug.
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    Measure Participants 66 65
    Number [Participants]
    65
    97%
    64
    97%
    8. Secondary Outcome
    Title Maximum Observed Serum Concentration (Cmax) of Antibody Drug Conjugate (ADC)
    Description Cmax of ADC was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    Pharmacokinetic Population (PKAS): (included all participants who were randomized to receive AGS-16C3F and had sufficient serum concentration data to facilitate derivation of at least 1 pharmacokinetic parameter, and for whom the time of dosing on the day of sampling was known) with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 59
    Cycle 1
    52.21
    (58.407)
    Cycle 4
    48.66
    (38.820)
    9. Secondary Outcome
    Title Mean Predose Serum Concentration (Ctrough) of ADC
    Description Ctrough of ADC was reported.
    Time Frame Predose on cycle 2, 3, 4, 6, 14, and 18 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population. Here, number of participants analyzed signifies participants who had quantifiable data.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 50
    Cycle 2
    3429.39
    (1402.334)
    Cycle 3
    5194.21
    (3602.012)
    Cycle 4
    7731.38
    (11171.906)
    Cycle 6
    5695.13
    (3899.222)
    Cycle 14
    8698.26
    (4239.327)
    Cycle 18
    7213.32
    (2082.501)
    10. Secondary Outcome
    Title Time to Maximum Observed Serum Concentration (Tmax) of ADC
    Description Tmax of ADC was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 59
    Cycle 1
    0.04
    Cycle 4
    0.05
    11. Secondary Outcome
    Title Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of ADC
    Description AUC (0 to 21) of ADC was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 52
    Cycle 1
    199.80
    (68.821)
    Cycle 4
    293.90
    (100.190)
    12. Secondary Outcome
    Title Terminal Elimination Half-life (t1/2) of ADC
    Description t1/2 of ADC was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 49
    Cycle 1
    6.93
    Cycle 4
    7.40
    13. Secondary Outcome
    Title Maximum Serum Concentration (Cmax) of Total Antibody (TAb)
    Description Cmax of TAb was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 59
    Cycle 1
    37.49
    (12.515)
    Cycle 4
    42.84
    (12.345)
    14. Secondary Outcome
    Title Mean Predose Serum Concnetration (Ctrough) of TAb
    Description Ctrough of TAb was reported.
    Time Frame Predose on cycle 2, 3, 4, 6, 14, and 18 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population. Here, number of participants analyzed signifies participants who had quantifiable data.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 50
    Cycle 2
    4966.69
    (2571.410)
    Cycle 3
    6943.11
    (3666.956)
    Cycle 4
    8660.94
    (4176.472)
    Cycle 6
    8777.51
    (4131.388)
    Cycle 14
    12491.90
    (6593.131)
    Cycle 18
    11726.15
    (5572.172)
    15. Secondary Outcome
    Title Time to Maximum Observed Serum Concentration (Tmax) of Tab
    Description Tmax of TAb was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 60
    Cycle 1
    0.05
    Cycle 4
    0.05
    16. Secondary Outcome
    Title Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of TAb
    Description AUC (0 to 21) of TAb was reporetd.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 48
    Cycle 1
    246.06
    (88.815)
    Cycle 4
    346.07
    (158.029)
    17. Secondary Outcome
    Title Terminal Elimination Half-life (t1/2) of Tab
    Description t1/2 of TAb was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 45
    Cycle 1
    8.70
    Cycle 4
    9.15
    18. Secondary Outcome
    Title Maximum Serum Concentration (Cmax) of Cysteine Adduct of Maleimidocaproyl Monomethyl Auristatin F (Cys-mcMMAF)
    Description Cmax of Cys-mcMMAF was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 64
    Cycle 1
    6.09
    (4.08)
    Cycle 4
    3.78
    (1.70)
    19. Secondary Outcome
    Title Mean Predose Serum Concnetration (Ctrough) of Cys-mcMMAF
    Description Ctrough of Cys-mcMMAF was reported.
    Time Frame Predose on cycle 2, 3, 4, 6, 14, and 18 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population. Here, number of participants analyzed signifies participants who had quantifiable data.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 1
    Cycle 6
    0.307
    (NA)
    20. Secondary Outcome
    Title Time to Maximum Observed Serum Concentration (Tmax) of Cys-mcMMAF
    Description Tmax of Cys-mcMMAF was reported.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 64
    Cycle 1
    0.207
    Cycle 4
    0.208
    21. Secondary Outcome
    Title Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of Cys-mcMMAF
    Description AUC (0 to 21) of Cys-mcMMAF was reporetd.
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point. Not calculated at cycle 4 due to an insufficient number of participants.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 21
    Cycle 1
    9.61
    (10.3)
    22. Secondary Outcome
    Title Terminal Elimination Half-life (t1/2) of Cys-mcMMAF
    Description t1/2 of Cys-mcMMAF was reported
    Time Frame Predose, end of infusion, 4, 24, 72, 336 hours postdose of cycle 1 and cycle 4 (each cycle is 21 days)

    Outcome Measure Data

    Analysis Population Description
    PKAS Population with available data at specified time point. Not calculated at cycle 4 due to an insufficient number of samples.
    Arm/Group Title AGS-16C3F
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion.
    Measure Participants 1
    Cycle 1
    2.72

    Adverse Events

    Time Frame From first dose up to 53 months
    Adverse Event Reporting Description All-cause mortality data was reported for FAS population. Serious and non-serious adverse events were reported for safety population.
    Arm/Group Title AGS-16C3F Axitinib
    Arm/Group Description Participants received 1.8 mg/kg of AGS-16C3F once every three weeks by single (60-minute) IV infusion. Participants received axitinib at a starting dose of 5 mg orally twice daily and then adjusted to 2 to 10 mg orally twice daily, as defined in the product label and per local institutional guidelines.
    All Cause Mortality
    AGS-16C3F Axitinib
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 36/67 (53.7%) 44/66 (66.7%)
    Serious Adverse Events
    AGS-16C3F Axitinib
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 26/66 (39.4%) 31/65 (47.7%)
    Blood and lymphatic system disorders
    Anaemia 1/66 (1.5%) 1 0/65 (0%) 0
    Iron deficiency anaemia 1/66 (1.5%) 1 0/65 (0%) 0
    Thrombocytopenia 2/66 (3%) 2 0/65 (0%) 0
    Cardiac disorders
    Angina pectoris 1/66 (1.5%) 2 0/65 (0%) 0
    Angina unstable 1/66 (1.5%) 1 0/65 (0%) 0
    Aortic valve disease 1/66 (1.5%) 1 0/65 (0%) 0
    Cardiac arrest 1/66 (1.5%) 1 1/65 (1.5%) 1
    Cardiac tamponade 0/66 (0%) 0 1/65 (1.5%) 1
    Cardio-respiratory arrest 0/66 (0%) 0 1/65 (1.5%) 1
    Pericardial effusion 0/66 (0%) 0 1/65 (1.5%) 1
    Sinus tachycardia 0/66 (0%) 0 1/65 (1.5%) 1
    Endocrine disorders
    Adrenal insufficiency 1/66 (1.5%) 1 0/65 (0%) 0
    Gastrointestinal disorders
    Abdominal hernia 0/66 (0%) 0 1/65 (1.5%) 1
    Abdominal pain 3/66 (4.5%) 4 0/65 (0%) 0
    Abdominal pain upper 0/66 (0%) 0 1/65 (1.5%) 1
    Diarrhoea 1/66 (1.5%) 1 2/65 (3.1%) 2
    Gastrointestinal haemorrhage 1/66 (1.5%) 2 0/65 (0%) 0
    Inguinal hernia 0/66 (0%) 0 1/65 (1.5%) 1
    Pancreatitis 0/66 (0%) 0 1/65 (1.5%) 1
    Pneumoperitoneum 0/66 (0%) 0 1/65 (1.5%) 1
    Small intestinal haemorrhage 1/66 (1.5%) 1 0/65 (0%) 0
    Small intestinal obstruction 0/66 (0%) 0 1/65 (1.5%) 1
    General disorders
    Asthenia 0/66 (0%) 0 1/65 (1.5%) 1
    Device failure 0/66 (0%) 0 1/65 (1.5%) 1
    Fatigue 0/66 (0%) 0 1/65 (1.5%) 1
    Influenza like illness 1/66 (1.5%) 1 0/65 (0%) 0
    Non-cardiac chest pain 1/66 (1.5%) 1 0/65 (0%) 0
    Pyrexia 1/66 (1.5%) 1 0/65 (0%) 0
    Hepatobiliary disorders
    Cholecystitis 0/66 (0%) 0 1/65 (1.5%) 1
    Hepatic failure 1/66 (1.5%) 1 0/65 (0%) 0
    Infections and infestations
    Clostridium difficile infection 1/66 (1.5%) 1 0/65 (0%) 0
    Osteomyelitis 0/66 (0%) 0 1/65 (1.5%) 1
    Pneumonia 0/66 (0%) 0 2/65 (3.1%) 2
    Sepsis 0/66 (0%) 0 1/65 (1.5%) 1
    Sinusitis 0/66 (0%) 0 1/65 (1.5%) 1
    Wound infection staphylococcal 0/66 (0%) 0 1/65 (1.5%) 1
    Injury, poisoning and procedural complications
    Fall 0/66 (0%) 0 1/65 (1.5%) 1
    Hip fracture 0/66 (0%) 0 1/65 (1.5%) 1
    Humerus fracture 1/66 (1.5%) 1 0/65 (0%) 0
    Infusion related reaction 1/66 (1.5%) 1 0/65 (0%) 0
    Post procedural haematoma 0/66 (0%) 0 1/65 (1.5%) 1
    Investigations
    Liver function test abnormal 1/66 (1.5%) 1 0/65 (0%) 0
    Platelet count decreased 1/66 (1.5%) 1 0/65 (0%) 0
    Transaminases increased 1/66 (1.5%) 1 0/65 (0%) 0
    Metabolism and nutrition disorders
    Dehydration 0/66 (0%) 0 4/65 (6.2%) 4
    Failure to thrive 1/66 (1.5%) 1 0/65 (0%) 0
    Hypercalcaemia 0/66 (0%) 0 1/65 (1.5%) 1
    Hyperglycaemia 1/66 (1.5%) 1 0/65 (0%) 0
    Hyponatraemia 1/66 (1.5%) 1 0/65 (0%) 0
    Malnutrition 0/66 (0%) 0 1/65 (1.5%) 1
    Musculoskeletal and connective tissue disorders
    Arthralgia 2/66 (3%) 2 0/65 (0%) 0
    Back pain 0/66 (0%) 0 2/65 (3.1%) 2
    Bone pain 0/66 (0%) 0 1/65 (1.5%) 1
    Cervical spinal stenosis 1/66 (1.5%) 1 0/65 (0%) 0
    Groin pain 1/66 (1.5%) 1 0/65 (0%) 0
    Haemarthrosis 0/66 (0%) 0 1/65 (1.5%) 1
    Muscle haemorrhage 1/66 (1.5%) 1 0/65 (0%) 0
    Pain in extremity 0/66 (0%) 0 2/65 (3.1%) 2
    Pathological fracture 0/66 (0%) 0 1/65 (1.5%) 1
    Soft tissue necrosis 0/66 (0%) 0 1/65 (1.5%) 1
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Metastases to central nervous system 1/66 (1.5%) 1 0/65 (0%) 0
    Metastatic pain 1/66 (1.5%) 1 0/65 (0%) 0
    Prostate cancer 0/66 (0%) 0 1/65 (1.5%) 1
    Nervous system disorders
    Altered state of consciousness 1/66 (1.5%) 1 0/65 (0%) 0
    Cauda equina syndrome 0/66 (0%) 0 1/65 (1.5%) 1
    Encephalopathy 1/66 (1.5%) 1 1/65 (1.5%) 1
    Haemorrhagic stroke 1/66 (1.5%) 1 0/65 (0%) 0
    Hemiparesis 1/66 (1.5%) 1 0/65 (0%) 0
    Lethargy 0/66 (0%) 0 1/65 (1.5%) 1
    Loss of consciousness 1/66 (1.5%) 1 0/65 (0%) 0
    Spinal cord compression 1/66 (1.5%) 2 0/65 (0%) 0
    Psychiatric disorders
    Confusional state 1/66 (1.5%) 1 2/65 (3.1%) 2
    Renal and urinary disorders
    Acute kidney injury 1/66 (1.5%) 1 2/65 (3.1%) 3
    Haematuria 1/66 (1.5%) 1 1/65 (1.5%) 1
    Reproductive system and breast disorders
    Vaginal haemorrhage 0/66 (0%) 0 1/65 (1.5%) 1
    Respiratory, thoracic and mediastinal disorders
    Dyspnoea 1/66 (1.5%) 1 2/65 (3.1%) 2
    Haemoptysis 0/66 (0%) 0 1/65 (1.5%) 1
    Hypoxia 1/66 (1.5%) 1 0/65 (0%) 0
    Pleural effusion 1/66 (1.5%) 1 1/65 (1.5%) 1
    Pneumothorax 0/66 (0%) 0 2/65 (3.1%) 3
    Pulmonary venous thrombosis 1/66 (1.5%) 1 0/65 (0%) 0
    Tracheal stenosis 0/66 (0%) 0 1/65 (1.5%) 1
    Surgical and medical procedures
    Craniotomy 0/66 (0%) 0 1/65 (1.5%) 1
    Vascular disorders
    Deep vein thrombosis 0/66 (0%) 0 1/65 (1.5%) 2
    Haematoma 1/66 (1.5%) 1 0/65 (0%) 0
    Hypotension 0/66 (0%) 0 1/65 (1.5%) 1
    Ischaemia 0/66 (0%) 0 1/65 (1.5%) 1
    Peripheral ischaemia 0/66 (0%) 0 1/65 (1.5%) 1
    Other (Not Including Serious) Adverse Events
    AGS-16C3F Axitinib
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 65/66 (98.5%) 64/65 (98.5%)
    Blood and lymphatic system disorders
    Anaemia 15/66 (22.7%) 35 4/65 (6.2%) 7
    Thrombocytopenia 5/66 (7.6%) 9 0/65 (0%) 0
    Endocrine disorders
    Hypothyroidism 1/66 (1.5%) 1 7/65 (10.8%) 8
    Eye disorders
    Diplopia 4/66 (6.1%) 4 0/65 (0%) 0
    Dry eye 14/66 (21.2%) 18 6/65 (9.2%) 6
    Eye pain 7/66 (10.6%) 7 0/65 (0%) 0
    Photophobia 5/66 (7.6%) 5 0/65 (0%) 0
    Vision blurred 26/66 (39.4%) 38 7/65 (10.8%) 8
    Gastrointestinal disorders
    Abdominal distension 4/66 (6.1%) 4 5/65 (7.7%) 5
    Abdominal pain 12/66 (18.2%) 20 10/65 (15.4%) 12
    Constipation 16/66 (24.2%) 16 21/65 (32.3%) 25
    Diarrhoea 12/66 (18.2%) 18 31/65 (47.7%) 70
    Dry mouth 6/66 (9.1%) 8 6/65 (9.2%) 10
    Dyspepsia 4/66 (6.1%) 6 7/65 (10.8%) 10
    Dysphagia 2/66 (3%) 2 5/65 (7.7%) 5
    Gastrooesophageal reflux disease 1/66 (1.5%) 2 7/65 (10.8%) 8
    Nausea 31/66 (47%) 56 26/65 (40%) 40
    Oral pain 0/66 (0%) 0 5/65 (7.7%) 8
    Stomatitis 2/66 (3%) 2 15/65 (23.1%) 23
    Vomiting 18/66 (27.3%) 25 22/65 (33.8%) 38
    General disorders
    Asthenia 7/66 (10.6%) 8 5/65 (7.7%) 7
    Chills 15/66 (22.7%) 29 2/65 (3.1%) 4
    Fatigue 36/66 (54.5%) 48 37/65 (56.9%) 67
    Influenza like illness 2/66 (3%) 2 5/65 (7.7%) 5
    Mucosal inflammation 1/66 (1.5%) 1 5/65 (7.7%) 9
    Non-cardiac chest pain 3/66 (4.5%) 3 4/65 (6.2%) 4
    Oedema peripheral 11/66 (16.7%) 13 7/65 (10.8%) 10
    Pain 1/66 (1.5%) 1 4/65 (6.2%) 4
    Pyrexia 9/66 (13.6%) 18 1/65 (1.5%) 1
    Infections and infestations
    Pneumonia 1/66 (1.5%) 1 5/65 (7.7%) 5
    Upper respiratory tract infection 4/66 (6.1%) 5 4/65 (6.2%) 4
    Injury, poisoning and procedural complications
    Contusion 5/66 (7.6%) 7 1/65 (1.5%) 1
    Fall 5/66 (7.6%) 7 1/65 (1.5%) 1
    Infusion related reaction 7/66 (10.6%) 16 0/65 (0%) 0
    Investigations
    Alanine aminotransferase increased 4/66 (6.1%) 4 3/65 (4.6%) 5
    Aspartate aminotransferase increased 5/66 (7.6%) 8 5/65 (7.7%) 7
    Blood bilirubin increased 4/66 (6.1%) 10 2/65 (3.1%) 3
    Blood creatinine increased 5/66 (7.6%) 8 5/65 (7.7%) 10
    Blood thyroid stimulating hormone increased 0/66 (0%) 0 5/65 (7.7%) 5
    Platelet count decreased 5/66 (7.6%) 9 2/65 (3.1%) 2
    Weight decreased 7/66 (10.6%) 13 19/65 (29.2%) 30
    Metabolism and nutrition disorders
    Decreased appetite 14/66 (21.2%) 17 24/65 (36.9%) 45
    Dehydration 6/66 (9.1%) 7 11/65 (16.9%) 15
    Hypercalcaemia 5/66 (7.6%) 6 3/65 (4.6%) 4
    Hypomagnesaemia 2/66 (3%) 2 4/65 (6.2%) 5
    Hyponatraemia 3/66 (4.5%) 6 5/65 (7.7%) 5
    Hypophosphataemia 1/66 (1.5%) 1 4/65 (6.2%) 6
    Musculoskeletal and connective tissue disorders
    Arthralgia 10/66 (15.2%) 13 9/65 (13.8%) 12
    Back pain 11/66 (16.7%) 23 24/65 (36.9%) 27
    Flank pain 3/66 (4.5%) 3 7/65 (10.8%) 7
    Groin pain 4/66 (6.1%) 4 2/65 (3.1%) 2
    Muscular weakness 5/66 (7.6%) 5 10/65 (15.4%) 12
    Musculoskeletal chest pain 6/66 (9.1%) 8 6/65 (9.2%) 7
    Musculoskeletal pain 11/66 (16.7%) 13 11/65 (16.9%) 13
    Neck pain 3/66 (4.5%) 3 7/65 (10.8%) 12
    Pain in extremity 6/66 (9.1%) 8 14/65 (21.5%) 22
    Nervous system disorders
    Dizziness 14/66 (21.2%) 18 12/65 (18.5%) 16
    Dysgeusia 3/66 (4.5%) 3 10/65 (15.4%) 11
    Headache 16/66 (24.2%) 20 16/65 (24.6%) 23
    Memory impairment 4/66 (6.1%) 4 0/65 (0%) 0
    Paraesthesia 2/66 (3%) 3 4/65 (6.2%) 4
    Peripheral sensory neuropathy 5/66 (7.6%) 6 2/65 (3.1%) 2
    Tremor 4/66 (6.1%) 4 0/65 (0%) 0
    Psychiatric disorders
    Anxiety 4/66 (6.1%) 4 2/65 (3.1%) 2
    Confusional state 4/66 (6.1%) 6 5/65 (7.7%) 7
    Insomnia 6/66 (9.1%) 6 7/65 (10.8%) 7
    Renal and urinary disorders
    Pollakiuria 1/66 (1.5%) 1 4/65 (6.2%) 4
    Proteinuria 1/66 (1.5%) 1 5/65 (7.7%) 10
    Respiratory, thoracic and mediastinal disorders
    Cough 17/66 (25.8%) 19 15/65 (23.1%) 20
    Dysphonia 1/66 (1.5%) 1 25/65 (38.5%) 26
    Dyspnoea 13/66 (19.7%) 19 18/65 (27.7%) 21
    Epistaxis 14/66 (21.2%) 23 5/65 (7.7%) 7
    Nasal congestion 1/66 (1.5%) 1 5/65 (7.7%) 7
    Oropharyngeal pain 4/66 (6.1%) 4 6/65 (9.2%) 7
    Pleural effusion 4/66 (6.1%) 8 7/65 (10.8%) 10
    Skin and subcutaneous tissue disorders
    Decubitus ulcer 1/66 (1.5%) 1 4/65 (6.2%) 4
    Night sweats 5/66 (7.6%) 5 2/65 (3.1%) 2
    Palmar-plantar erythrodysaesthesia syndrome 0/66 (0%) 0 14/65 (21.5%) 38
    Pruritus 11/66 (16.7%) 12 3/65 (4.6%) 3
    Rash 6/66 (9.1%) 7 4/65 (6.2%) 4
    Rash maculo-papular 5/66 (7.6%) 6 3/65 (4.6%) 5
    Vascular disorders
    Hypertension 4/66 (6.1%) 10 27/65 (41.5%) 48
    Hypotension 4/66 (6.1%) 6 6/65 (9.2%) 7

    Limitations/Caveats

    [Not Specified]

    More Information

    Certain Agreements

    Principal Investigators are NOT employed by the organization sponsoring the study.

    Institute and/or Principal Investigator may publish trial data generated at their specific study site after Sponsor publication of the multi-center data. Sponsor must receive a site's manuscript prior to publication for review and comment as specified in the Investigator Agreement.

    Results Point of Contact

    Name/Title Clinical Trial Disclosure
    Organization Astellas Pharma Global Development, Inc.
    Phone 800-888-7704
    Email astellas.resultsdisclosure@astellas.com
    Responsible Party:
    Astellas Pharma Global Development, Inc.
    ClinicalTrials.gov Identifier:
    NCT02639182
    Other Study ID Numbers:
    • AGS-16C3F-15-3
    First Posted:
    Dec 24, 2015
    Last Update Posted:
    Nov 10, 2021
    Last Verified:
    Nov 1, 2021