search
Back to results

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

Primary Purpose

Metastatic Renal Cell Carcinoma

Status
Completed
Phase
Phase 2
Locations
International
Study Type
Interventional
Intervention
AGS-16C3F
Axitinib
Sponsored by
Astellas Pharma Global Development, Inc.
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Metastatic Renal Cell Carcinoma focused on measuring Axitinib, Metastatic Renal Cell Carcinoma, AGS-16C3F

Eligibility Criteria

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

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.

Sites / Locations

  • Site US01026
  • Site US01008
  • Site US01007
  • Site US01020
  • Site US01019
  • Site US01010
  • Site US01023
  • Site US01002
  • Site US01004
  • Site US01013
  • Site US01012
  • Site US01006
  • Site US01022
  • Site US01017
  • Site US01021
  • Site US01011
  • Site US01003
  • Site US01014
  • Site US01001
  • Site US01009
  • Site CA02006
  • Site CA02004
  • Site CA02005
  • Site CA02001
  • Site CA02002
  • Site CA02008

Arms of the Study

Arm 1

Arm 2

Arm Type

Experimental

Active Comparator

Arm Label

AGS-16C3F

Axitinib

Arm Description

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

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.

Outcomes

Primary Outcome Measures

Progression Free Survival (PFS) as Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Investigator Radiologic Review
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

PFS Per RECIST v1.1 as Assessed by Blinded Central Radiology Assessment
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.
Objective Response Rate (ORR) Based on the Investigator's Radiographic Assessment
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.
Duration of Response (DOR) Based on the Investigator's Radiographic Assessment
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.
Overall Survival (OS)
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.
Disease Control Rate (DCR) Based on the Investigator's Radiographic Assessment
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.
Number of Participants With Adverse Events
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.
Maximum Observed Serum Concentration (Cmax) of Antibody Drug Conjugate (ADC)
Cmax of ADC was reported.
Mean Predose Serum Concentration (Ctrough) of ADC
Ctrough of ADC was reported.
Time to Maximum Observed Serum Concentration (Tmax) of ADC
Tmax of ADC was reported.
Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of ADC
AUC (0 to 21) of ADC was reported.
Terminal Elimination Half-life (t1/2) of ADC
t1/2 of ADC was reported.
Maximum Serum Concentration (Cmax) of Total Antibody (TAb)
Cmax of TAb was reported.
Mean Predose Serum Concnetration (Ctrough) of TAb
Ctrough of TAb was reported.
Time to Maximum Observed Serum Concentration (Tmax) of Tab
Tmax of TAb was reported.
Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of TAb
AUC (0 to 21) of TAb was reporetd.
Terminal Elimination Half-life (t1/2) of Tab
t1/2 of TAb was reported.
Maximum Serum Concentration (Cmax) of Cysteine Adduct of Maleimidocaproyl Monomethyl Auristatin F (Cys-mcMMAF)
Cmax of Cys-mcMMAF was reported.
Mean Predose Serum Concnetration (Ctrough) of Cys-mcMMAF
Ctrough of Cys-mcMMAF was reported.
Time to Maximum Observed Serum Concentration (Tmax) of Cys-mcMMAF
Tmax of Cys-mcMMAF was reported.
Area Under the Concentration Versus Time Curve From Time Zero to 21days (AUC0 to 21) of Cys-mcMMAF
AUC (0 to 21) of Cys-mcMMAF was reporetd.
Terminal Elimination Half-life (t1/2) of Cys-mcMMAF
t1/2 of Cys-mcMMAF was reported

Full Information

First Posted
December 21, 2015
Last Updated
November 9, 2021
Sponsor
Astellas Pharma Global Development, Inc.
search

1. Study Identification

Unique Protocol Identification Number
NCT02639182
Brief Title
A Study of AGS-16C3F vs. Axitinib in Metastatic Renal Cell Carcinoma
Official Title
A Multi-Center, Open Label, Randomized Phase 2 Study of AGS-16C3F vs. Axitinib in Metastatic Renal Cell Carcinoma
Study Type
Interventional

2. Study Status

Record Verification Date
November 2021
Overall Recruitment Status
Completed
Study Start Date
May 3, 2016 (Actual)
Primary Completion Date
October 2, 2020 (Actual)
Study Completion Date
October 2, 2020 (Actual)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Astellas Pharma Global Development, Inc.

4. Oversight

Studies a U.S. FDA-regulated Drug Product
Yes
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. 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.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Metastatic Renal Cell Carcinoma
Keywords
Axitinib, Metastatic Renal Cell Carcinoma, AGS-16C3F

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 2
Interventional Study Model
Parallel Assignment
Masking
None (Open Label)
Allocation
Randomized
Enrollment
133 (Actual)

8. Arms, Groups, and Interventions

Arm Title
AGS-16C3F
Arm Type
Experimental
Arm Description
Participants received 1.8 milligram per kilogram (mg/kg) of AGS-16C3F once every three weeks by single intravenous (IV) infusion.
Arm Title
Axitinib
Arm Type
Active Comparator
Arm Description
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.
Intervention Type
Drug
Intervention Name(s)
AGS-16C3F
Intervention Description
Intravenous (IV) infusion
Intervention Type
Drug
Intervention Name(s)
Axitinib
Other Intervention Name(s)
Inlyta®
Intervention Description
Oral
Primary Outcome Measure Information:
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)
Secondary Outcome Measure Information:
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)
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)
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)
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)
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)
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
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)
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)
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)
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)
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)
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)
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)
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)
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)
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)
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)
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)
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)
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)
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)

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
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: Absolute neutrophil count (ANC) ≥ 1.5 x 10 9/L Platelet count ≥ 100 x 10 9/L 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: Aspartate aminotransferase (AST) and Alanine aminotransferase (ALT) ≤ 2.5 x ULN or ≤ 5x ULN if known liver metastases 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: post-menopausal (defined as at least 1 year without any menses) prior to Screening, or documented surgically sterile Or, if of childbearing potential, Agree not to try to become pregnant during the study and for 6 months after the final study drug administration 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.
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Associate Medical Director
Organizational Affiliation
Astellas Pharma Global Development, Inc.
Official's Role
Study Director
Facility Information:
Facility Name
Site US01026
City
Tucson
State/Province
Arizona
ZIP/Postal Code
85719
Country
United States
Facility Name
Site US01008
City
La Jolla
State/Province
California
ZIP/Postal Code
92093
Country
United States
Facility Name
Site US01007
City
Los Angeles
State/Province
California
ZIP/Postal Code
90033
Country
United States
Facility Name
Site US01020
City
Los Angeles
State/Province
California
ZIP/Postal Code
90095
Country
United States
Facility Name
Site US01019
City
Palo Alto
State/Province
California
ZIP/Postal Code
94305
Country
United States
Facility Name
Site US01010
City
Atlanta
State/Province
Georgia
ZIP/Postal Code
30322
Country
United States
Facility Name
Site US01023
City
Baltimore
State/Province
Maryland
ZIP/Postal Code
21201
Country
United States
Facility Name
Site US01002
City
Boston
State/Province
Massachusetts
ZIP/Postal Code
02215
Country
United States
Facility Name
Site US01004
City
Ann Arbor
State/Province
Michigan
ZIP/Postal Code
48109
Country
United States
Facility Name
Site US01013
City
Detroit
State/Province
Michigan
ZIP/Postal Code
48202
Country
United States
Facility Name
Site US01012
City
Omaha
State/Province
Nebraska
ZIP/Postal Code
68130
Country
United States
Facility Name
Site US01006
City
Buffalo
State/Province
New York
ZIP/Postal Code
14263
Country
United States
Facility Name
Site US01022
City
Durham
State/Province
North Carolina
ZIP/Postal Code
27710
Country
United States
Facility Name
Site US01017
City
Portland
State/Province
Oregon
ZIP/Postal Code
97213
Country
United States
Facility Name
Site US01021
City
Pittsburgh
State/Province
Pennsylvania
ZIP/Postal Code
15232
Country
United States
Facility Name
Site US01011
City
Charleston
State/Province
South Carolina
ZIP/Postal Code
29425
Country
United States
Facility Name
Site US01003
City
Houston
State/Province
Texas
ZIP/Postal Code
77030
Country
United States
Facility Name
Site US01014
City
Temple
State/Province
Texas
ZIP/Postal Code
76508
Country
United States
Facility Name
Site US01001
City
Seattle
State/Province
Washington
ZIP/Postal Code
98109
Country
United States
Facility Name
Site US01009
City
Milwaukee
State/Province
Wisconsin
ZIP/Postal Code
53226
Country
United States
Facility Name
Site CA02006
City
Calgary
State/Province
Alberta
ZIP/Postal Code
T2N 4N2
Country
Canada
Facility Name
Site CA02004
City
Edmonton
State/Province
Alberta
ZIP/Postal Code
T6G 1Z2
Country
Canada
Facility Name
Site CA02005
City
Kelowna
State/Province
British Columbia
ZIP/Postal Code
V1Y 5L3
Country
Canada
Facility Name
Site CA02001
City
Vancouver
State/Province
British Columbia
ZIP/Postal Code
V5Z 4E6
Country
Canada
Facility Name
Site CA02002
City
Hamilton
State/Province
Ontario
ZIP/Postal Code
L8V 5C2
Country
Canada
Facility Name
Site CA02008
City
London
State/Province
Ontario
ZIP/Postal Code
N6A 5W9
Country
Canada

12. IPD Sharing Statement

Plan to Share IPD
Yes
IPD Sharing Plan Description
Access to anonymized individual participant level data collected during the study, in addition to study-related supporting documentation, is planned for studies conducted with approved product indications and formulations, as well as compounds terminated during development. Studies conducted with product indications or formulations that remain active in development are assessed after study completion to determine if Individual Participant Data can be shared. Conditions and exceptions are described under the Sponsor Specific Details for Astellas on www.clinicalstudydatarequest.com.
IPD Sharing Time Frame
Access to participant level data is offered to researchers after publication of the primary manuscript (if applicable) and is available as long as Astellas has legal authority to provide the data.
IPD Sharing Access Criteria
Researchers must submit a proposal to conduct a scientifically relevant analysis of the study data. The research proposal is reviewed by an Independent Research Panel. If the proposal is approved, access to the study data is provided in a secure data sharing environment after receipt of a signed Data Sharing Agreement.
IPD Sharing URL
https://www.clinicalstudydatarequest.com/
Links:
URL
https://www.clinicaltrials.astellas.com/study/?pid=AGS-16C3F-15-3
Description
Link to results and other applicable study documents on the Astellas Clinical Trials website
URL
https://www.trialsummaries.com/Study/StudyDetails?id=14554&tenant=MT_AST_9011
Description
Link to plain language summary of the study on the Trial Results Summaries website

Learn more about this trial

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

We'll reach out to this number within 24 hrs