search
Back to results

A Study to Assess the Antitumor Activity, Safety, Pharmacokinetics and Biomarkers of Zolbetuximab (IMAB362) in Participants With Claudin (CLDN) 18.2 Positive, Metastatic or Advanced Unresectable Gastric and Gastroesophageal Junction (GEJ) Adenocarcinoma (ILUSTRO)

Primary Purpose

Pharmacokinetics of Zolbetuximab, Gastric Cancer, Gastro-esophageal Junction (GEJ) Cancer

Status
Recruiting
Phase
Phase 2
Locations
International
Study Type
Interventional
Intervention
zolbetuximab
oxaliplatin
leucovorin
fluorouracil
Pembrolizumab
folinic acid
nivolumab
Sponsored by
Astellas Pharma Global Development, Inc.
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Pharmacokinetics of Zolbetuximab focused on measuring zolbetuximab, oxaliplatin, fluorouracil, claudiximab, IMAB362, Gastro-Esophageal Junction cancer, Pembrolizumab, Gastric cancer

Eligibility Criteria

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

Inclusion Criteria:

  • Female subject eligible to participate if she is not pregnant and at least one of the following conditions applies:

    • Not a woman of child-bearing potential (WOCBP) OR
    • WOCBP who agrees to follow the contraceptive guidance throughout the treatment period and for at least 9 months after the final oxaliplatin administration and 6 months after the final administration of all other study drugs.
  • 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 agree not to donate ova starting at screening and throughout the study period, and for 9 months after the final oxaliplatin administration and 6 months after the final administration of all other study drugs.
  • A sexually active male subject with a female partner(s) who is of child-bearing potential must agree to use contraception during the treatment period and for at least 6 months after the final study drug administration.
  • Male subject must agree not to donate sperm starting at screening and throughout the study period, and for 6 months after the final study drug administration.
  • Male subject with a pregnant or breastfeeding partner(s) must agree to remain abstinent or use a condom for the duration of the pregnancy or time partner is breastfeeding throughout the study period and for 6 months after the final study drug administration.
  • Subject has histologically confirmed gastric or GEJ adenocarcinoma.
  • Subject has radiographically-confirmed, locally advanced, unresectable or metastatic disease within 28 days prior to the first dose of study treatment.
  • Subject's tumor is positive for CLDN18.2 expression.
  • Subject agrees to not participate in another interventional study while on treatment.
  • Subject has ECOG performance status 0 to 1.
  • Subject has predicted life expectancy ≥ 12 weeks.
  • Subject must meet all of the following criteria based on the centrally or locally analyzed laboratory tests collected within 14 days prior to the first dose of study treatment. In case of multiple central laboratory data within this period, the most recent data should be used.

    • Hemoglobin (Hgb) ≥ 9 g/dL (transfusion is allowed, but post-transfusion Hgb [24 hours or later following transfusion] must be ≥ 9 g/dL)
    • Absolute neutrophil count (ANC) ≥ 1.5 × 109/L
    • Platelets ≥ 100 × 10^9/L
    • Albumin ≥ 2.5 g/dL
    • Total bilirubin ≤ 1.5 × upper limit of normal (ULN)
    • Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) ≤ 2.5 × ULN in subjects without liver metastases (≤ 5 × ULN if liver metastases are present)
    • Estimated creatinine clearance ≥ 30 mL/min
    • Prothrombin time/international normalized ratio and partial thromboplastin time ≤ 1.5 × ULN (except for subjects receiving anticoagulation therapy)

Specific to Cohort 1A:

  • Subject has measurable disease according to RECIST 1.1 within 28 days prior to the first dose of study treatment per investigator assessment. For subjects with only 1 evaluable lesion and prior radiotherapy ≤ 3 months before enrollment, the lesion must either be outside the field of prior radiotherapy or must have documented progression following radiation therapy.
  • Subject has disease progression on or after at least 2 prior regimens for their advanced disease, including fluoropyrimidine and platinum-containing chemotherapy, and if appropriate, HER2/neu-targeted therapy and all associated side effects have resolved to grade 1 or less.
  • Subject must have an additional available tumor specimen collected within 3 months prior to the first dose of study treatment.
  • Subject must be an appropriate candidate for a tumor biopsy and is amenable to undergo a tumor biopsy during the screening period (if applicable) and treatment period as indicated in the Schedule of Assessments.

Specific to Cohort 2:

  • Subject has measurable disease according to RECIST 1.1 within 28 days prior to the first dose of study treatment per investigator assessment. For subjects with only 1 evaluable lesion and prior radiotherapy ≤ 3 months before enrollment, the lesion must either be outside the field of prior radiotherapy or must have documented progression following radiation therapy.
  • Subject has not received prior systemic anti-cancer therapy for their advanced disease (subject may have received neoadjuvant and/or fluorouracil-containing adjuvant chemotherapy as long as it has been completed ≥ 6 months before the first dose of study treatment).
  • Subject has a gastric or GEJ tumor that is HER2-negative as determined by local or central testing.
  • Subject must have an additional available tumor specimen collected within 3 months prior to the first dose of study treatment.
  • Subject must be an appropriate candidate for a tumor biopsy and is amenable to undergo a tumor biopsy during the screening period (if applicable) and treatment period as indicated in the Schedule of Assessments.

Specific to Cohort 3A:

  • Subject has radiologically evaluable disease (measurable and/or non-measurable) according to RECIST 1.1, per local assessment, ≤ 28 days prior to the first dose of study treatment. For subjects with only 1 evaluable lesion and prior radiotherapy ≤ 3 months before enrollment, the lesion must either be outside the field of prior radiotherapy or must have documented progression following radiation therapy.
  • Subject has disease progression on or after at least 2 prior regimens for their advanced disease, including fluoropyrimidine and platinum-containing chemotherapy, and if appropriate, HER2/neu-targeted therapy.
  • Subject has not received prior checkpoint inhibitor therapy.

Specific to Cohort 4A and 4B:

  • Subject has radiologically evaluable disease.
  • Subject has not received prior systemic anti-cancer therapy for their advanced disease.
  • Subject has a gastric or GEJ tumor that is HER2-negative as determined by local or central testing.
  • Subject has not received prior checkpoint inhibitor therapy.

Specific to Cohort 4B Only:

  • Subject must have an additional available tumor specimen collected within 3 months prior to the first dose of study treatment.
  • Subject must be an appropriate candidate for a tumor biopsy and is amenable to undergo a tumor biopsy during the screening period (if applicable) and treatment period.
  • Subject has a tumor that is PD-L1 positive.

Exclusion Criteria:

  • Subject has had prior severe allergic reaction or intolerance to known ingredients of zolbetuximab or other monoclonal antibodies, including humanized or chimeric antibodies.
  • Subject has known immediate or delayed hypersensitivity or contraindication to any component of study treatment.
  • Subject has received other investigational agents or devices concurrently or within 28 days prior to first dose of study treatment.
  • Subject has received systemic immunosuppressive therapy, including systemic corticosteroids 14 days prior to first dose of study treatment.
  • Subject has a complete gastric outlet syndrome or a partial gastric outlet syndrome with persistent recurrent vomiting.
  • Subject has significant gastric bleeding and/or untreated gastric ulcers that would preclude the subject from participation.
  • Subject has history of central nervous system metastases and/or carcinomatous meningitis from gastric/GEJ cancer.
  • Subject has a known history of a positive test for human immunodeficiency virus (HIV) infection or known active hepatitis B (positive hepatitis B surface antigen [HBsAg]) or hepatitis C infection.
  • Subject has had within 6 months prior to first dose of study treatment any of the following: unstable angina, myocardial infarction, ventricular arrhythmia requiring intervention or hospitalization for heart failure.
  • Subject has active infection requiring systemic therapy that has not completely resolved within 7 days prior to the start of study treatment.
  • Subject has active autoimmune disease that has required systemic treatment within the past 3 months prior to the start of study treatment.
  • Subject has a clinically significant disease or co-morbidity that may adversely affect the safe delivery of treatment within this study or make the subject unsuitable for study participation.
  • Subject has psychiatric illness or social situations that would preclude study compliance.
  • Subject has had a major surgical procedure ≤ 28 days before start of study treatment.
  • Subject is without complete recovery from a major surgical procedure ≤ 14 days before start of study treatment

    • Subject has received radiotherapy for locally advanced unresectable or metastatic gastric or GEJ adenocarcinoma ≤ 14 days (Cohorts 1 and 3A) and ≤ 28 days (Cohorts 2 and 4A or 4B) prior to start of study treatment and has NOT recovered from any related toxicity.
    • Subject has another malignancy, for which treatment is required.
  • Cohort 2 and 4 Only, subject has any of the following:

    • Prior severe allergic reaction or intolerance to any component of mFOLFOX6 chemotherapeutics in this study
    • Known dihydropyrimidine dehydrogenase deficiency (DPD).
    • Known peripheral neuropathy > Grade 1 (absence of deep tendon reflexes as the sole neurological abnormality does not render the subject ineligible).
    • Sinusoidal obstruction syndrome, formerly known as veno-occlusive disease, if present, should be stable or improving.
    • History of clinically significant ventricular arrhythmias.
    • QTc interval > 450 msec for male subjects; QTc interval > 470 msec for female subjects.
    • History or family history of congenital long QT syndrome.
    • Cardiac arrhythmias requiring anti-arrhythmic medications (Subjects with rate controlled atrial fibrillation for > 1 month prior to first dose of study treatment are eligible).
  • Cohorts 3A, 4A and 4B Only, subject has any of the following:

    • Subjects with ongoing or previous autoimmune disease or interstitial lung disease, active diverticulitis or gastrointestinal ulcerative disease, or solid organ or stem cell transplant (for Cohort 4) or other uncontrolled or clinically significant medical disorders.
    • Subjects with type 1 diabetes mellitus, endocrinopathies stably maintained on appropriate replacement therapy or skin disorders (e.g., vitiligo, psoriasis, or alopecia) not requiring systemic treatment are allowed.
    • Subject has known history of serious hypersensitivity reaction to a known ingredient of pembrolizumab or nivolumab.
  • Cohort 4B Only: Subject with known microsatellite instability-high or mismatch repair deficient tumors.

Sites / Locations

  • The Angeles Clinic and Research Institute
  • UCLA Medical Center
  • Georgetown Univ Hospital
  • University of ChicagoRecruiting
  • Indiana University Cancer Center
  • Mass General / North Shore CanRecruiting
  • Karmanos Cancer Institute
  • Memorial Sloan Kettering Cancer Center
  • Weill Cornell Medical College
  • Sanford Cancer Center
  • Virginia Cancer Specialists
  • Site FR33003Recruiting
  • Site FR33002Recruiting
  • Site FR33001Recruiting
  • Site FR33004Recruiting
  • Site IT39005Recruiting
  • Site IT39002
  • Site IT39004
  • Site IT39003
  • Site JP81001Recruiting
  • Site JP81002Recruiting
  • Site JP81003Recruiting
  • Site KR82002Recruiting
  • Site KR82001Recruiting
  • Site TW88601Recruiting
  • Site TW88602

Arms of the Study

Arm 1

Arm 2

Arm 3

Arm 4

Arm Type

Experimental

Experimental

Experimental

Experimental

Arm Label

zolbetuximab (Cohort 1A)

mFOLFOX6 plus zolbetuximab (Cohort 2)

Pembrolizumab plus zolbetuximab (Cohort 3A)

Zolbetuximab in combination with mFOLFOX6 and nivolumab (Cohort 4A/4B)

Arm Description

Participants will be treated with zolbetuximab on a 21-day cycle in which zolbetuximab will be administered as a single agent every 3 weeks until disease progression, toxicity requiring cessation, start of another anti-cancer treatment or other treatment discontinuation criteria are met.

Participants will be treated with zolbetuximab and mFOLFOX6 on a 42-day cycle in which zolbetuximab is administered on days 1 and 22, and mFOLFOX6 is administered on days 1, 15 and 29; however, for the first cycle, zolbetuximab will be administered on day 3 (instead of day 1) to allow for pharmacokinetic collection. Participants will receive up to 12 mFOLFOX6 treatments (4 cycles). Beginning at cycle 5, participants may continue on 5-FU and leucovorin or folinic acid along with zolbetuximab for the remainder of the study per investigator's discretion. mFOLFOX6 treatment includes oxaliplatin: intravenous [IV] infusion, leucovorin: IV infusion, fluorouracil bolus: IV bolus, fluorouracil infusion: continuous IV infusion.

Participants will be treated with zolbetuximab and pembrolizumab on a 21-day cycle. Loading dose of zolbetuximab will be administered at cycle 1, day 1 followed by maintenance dose of zolbetuximab once every 3 weeks (Q3W). Pembrolizumab will be administered to 3 to 6 subjects at a intravenously on day 1 of every 21-day cycle and will be infused 1 hour after the zolbetuximab infusion is completed. Tolerability and safety of zolbetuximab in combination with pembrolizumab will be evaluated during the 3-week dose-limiting toxicity (DLT) assessment period. If this cycle 1 dose is not tolerable, a lower dose of zolbetuximab in combination with pembrolizumab will subsequently be evaluated.

Participants will be treated with zolbetuximab and mFOLFOX6, nivolumab on a 42-day cycle. Cohort 4A: Loading dose of zolbetuximab in combination with nivolumab and mFOLFOX6 on cycle 1 day 1, followed by zolbetuximab in combination with nivolumab and mFOLFOX6 q2w [days 15 and 29] (1 cycle = 6 weeks). Tolerability and safety of zolbetuximab in combination with nivolumab, mFOLFOX6 will be evaluated during the 3-week DLT assessment period. If cycle 1 dose is not tolerable, a lower dose of dose zolbetuximab in combination with nivolumab and mFOLFOX6 will be subsequently evaluated. Cohort 4B: Subjects will be treated with the combination of zolbetuximab, mFOLFOX6 and nivolumab at the dose deemed tolerable in Cohort 4A. Subjects will receive up to 12 mFOLFOX6 treatments (4 cycles). For Cohorts 4A and 4B, beginning at cycle 5, subjects may continue on 5-FU and leucovorin or folinic acid along with zolbetuximab and nivolumab for the remainder of the study per investigator's discretion.

Outcomes

Primary Outcome Measures

Objective Response Rate (ORR) of zolbetuximab as a single agent by central review (Cohort 1)
The ORR is defined as the proportion of participants with complete or partial objective response based on Response Evaluation Criteria in Solid Tumors (RECIST) V1.1 (assessed by an independent review committee (IRC)).

Secondary Outcome Measures

Pharmacokinetics (PK) of zolbetuximab: Area Under the Concentration-time Curve from the Time of Dosing Extrapolated to Time Infinity (AUCinf) (Cohorts 1A, 2, 3A and 4)
AUCinf will be derived from the PK serum samples collected.
PK of zolbetuximab: Area Under the Concentration-time Curve from the Time of Dosing Extrapolated to Time Infinity Percentage (AUCinf (%extrap)) (Cohorts 1A, 2, 3A and 4)
AUCinf (%extrap) will be derived from the PK serum samples collected.
PK of zolbetuximab: Area Under the Concentration-time Curve from the Time of Dosing Until the Last Measurable Concentration (AUClast) (Cohorts 1A, 2, 3A and 4)
AUClast will be derived from the PK serum samples collected.
PK of zolbetuximab: Area Under the Concentration-Time Curve from the Time of Dosing to the Start of the Next Dosing Interval (AUCtau) (Cohorts 1A, 2, 3A and 4)
AUCtau will be derived from the PK serum samples collected.
PK of zolbetuximab: Maximum Concentration (Cmax) (Cohorts 1A, 2, 3A and 4)
Cmax will be derived from the PK serum samples collected.
PK of zolbetuximab: Concentration Immediately Prior to Dosing (Ctrough) (Cohorts 1A, 2, 3A and 4)
Ctrough will be derived from the PK serum samples collected.
PK of zolbetuximab: Time of Maximum Concentration (Tmax) (Cohorts 1A, 2, 3A and 4)
Tmax will be derived from the PK serum samples collected.
PK of zolbetuximab: Terminal Elimination Half-life (T1/2) (Cohorts 1A, 2, 3A and 4)
T1/2 will be derived from the PK serum samples collected.
PK of zolbetuximab: Time of the last measurable concentration (Tlast) (Cohorts 1A, 2, 3A and 4)
Tlast will be derived from the PK serum samples collected.
PK of zolbetuximab: Clearance (CL) (Cohorts 1A, 2, 3A and 4)
CL will be derived from the PK serum samples collected.
PK of zolbetuximab: Volume of Distribution During the Terminal Phase (Vz) (Cohorts 1A, 2, 3A and 4)
Vz will be derived from the PK serum samples collected.
PK of oxaliplatin: AUCinf (Cohort 2)
AUCinf will be derived from the PK plasma samples collected.
PK of oxaliplatin: AUCinf (%extrap) (Cohort 2)
AUCinf (%extrap) will be derived from the PK plasma samples collected.
PK of oxaliplatin: AUClast (Cohort 2)
AUClast will be derived from the PK plasma samples collected.
PK of oxaliplatin: Cmax (Cohort 2)
Cmax will be derived from the PK plasma samples collected.
PK of oxaliplatin: Tmax (Cohort 2)
Tmax will be derived from the PK plasma samples collected.
PK of oxaliplatin: T1/2 (Cohort 2)
T1/2 will be derived from the PK plasma samples collected.
PK of oxaliplatin: Tlast (Cohort 2)
Tlast will be derived from the PK plasma samples collected.
PK of oxaliplatin: CL (Cohort 2)
TL will be derived from the PK plasma samples collected.
PK of oxaliplatin: Vz (Cohort 2)
Vz will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): AUCinf (Cohort 2)
AUCinf will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): AUCinf (%extrap) (Cohort 2)
AUCinf (%extrap) will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): AUClast (Cohort 2)
AUClast will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): Cmax (Cohort 2)
Cmax will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): Tmax (Cohort 2)
Tmax will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): T1/2 (Cohort 2)
T1/2 will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): Tlast (Cohort 2)
Tlast will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): CL (Cohort 2)
CL will be derived from the PK plasma samples collected.
PK of fluorouracil bolus (5-FU): Vz (Cohort 2)
Vz will be derived from the PK plasma samples collected.
Safety and tolerability assessed by adverse events (AEs) (Cohorts 1A, 2, 3A and 4)
An AE is any untoward medical occurrence in a participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product.
Number of participants with electrocardiogram (ECG) abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Number of participants with potentially clinically significant ECG values.
Number of participants with vital signs abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Number of participants with potentially clinically significant vital sign values.
Number of participants with European Cooperative Oncology Group (ECOG) performance status abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Number of participants with potentially clinically significant ECOG performance status values. ECOG grades 0-5, where 0 = Fully active, able to carry on all pre-disease performance without restriction; 1 = Restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature, e.g., light house work, office work; 2 = Ambulatory and capable of all self-care but unable to carry out any work activities. Up and about more than 50% of waking hours; 3 = Capable of only limited self-care, confined to bed or chair more than 50% of waking hours; 4 = Completely disabled. Cannot carry on any self-care. Totally confined to bed or chair and 5 = Dead.
Number of participants with laboratory assessments abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Number of participants with potentially clinically significant laboratory values.
Number of anti-drug antibody (ADA) Positive Participants (Cohorts 1A, 2, 3A and 4)
Immunogenicity will be measured by the number of participants that are ADA positive.
Health Related Quality of Life (HRQoL) measured by the Quality of Life Questionnaire - Core Questionnaire (QLQ-C30) questionnaire (Cohorts 1A, 2, 3A and 4)
The EORTC-QLQ-C30 is a cancer-specific instrument consisting of 5 functional domain scales: physical, role, emotional, social and cognitive.
HRQoL measured by the Oesophago-Gastric Module (EORTC QLQ-OG-25) questionnaire (Cohorts 1A, 2, 3A and 4)
The EORTC-QLQ-OG25 instrument evaluates GC- and GEJC-specific symptoms such as stomach discomfort, difficulties eating and swallowing and indigestion.
HRQoL measured by the Global Pain (GP) questionnaire (Cohorts 1A, 2, 3A and 4)
The GP instrument is a single assessment of overall pain.
HRQoL measured by the EuroQOL Five Dimensions Questionnaire 5L (EQ-5D-5L) questionnaire (Cohorts 1A, 2, 3A and 4)
The EQ-5D-5L is a standardized instrument developed by the EuroQol Group for use as a generic, preference-based measure of health outcomes. The EQ-5D-5L is a 5-item self-reported measure of functioning and wellbeing, which assesses 5 dimensions of health, including mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension comprises 5 levels (no problems, slight problems, moderate problems, severe problems, extreme problems). A unique EQ-5D-5L health state is defined by combining 1 level from each of the 5 dimensions. This questionnaire also records the respondent's self-rated health status on a vertical graduated (0 = the worst health a participant can imagine to 100 = the best health a participant can imagine) visual analogue scale. Responses to the 5 items will also be converted to a weighted health state index (utility score) based on values derived from general population samples.
HRQoL measured by the Health Resource Utilization (HRU) questionnaire (Cohorts 1A, 2, 3A and 4)
Health resource utilization questionnaire to assess the number of office visits, hospital stays and other healthcare resource utilization that occur outside of the clinical trial.
Disease Control Rate (DCR) of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
DCR of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
DCR of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
DCR of zolbetuximab as a single agent by independent central reader (Cohort 1A)
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
DCR of zolbetuximab in combination with mFOLFOX6 by Independent central reader (Cohort 2)
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
Duration of Response (DOR) of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
DOR of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
DOR of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
DOR of zolbetuximab as a single agent by independent central reader (Cohort 1A)
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
DOR of zolbetuximab in combination with mFOLFOX6 by independent central reader (Cohort 2)
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
Progression Free Survival (PFS) of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
PFS of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
PFS of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
PFS of zolbetuximab as a single agent by independent central review (Cohort 1A)
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
PFS of zolbetuximab in combination with mFOLFOX6 by independent central review (Cohort 2)
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
ORR of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
ORR of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
ORR of zolbetuximab in combination with pembrolizumab by investigator assessment (Cohort 3A)
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
ORR of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
ORR of zolbetuximab in combination with mFOLFOX6 by independent central reader (Cohort 2)
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
ORR of zolbetuximab in combination with pembrolizumab by independent central reader (Cohort 3A)
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
Overall Survival (OS) of zolbetuximab as a single agent (Cohort 1A)
OS is defined as the time from the date of treatment start until the documented date of death from any cause.
OS of zolbetuximab in combination with mFOLFOX6 and nivolumab (Cohort 4B)
OS is defined as the time from the date of treatment start until the documented date of death from any cause.

Full Information

First Posted
March 29, 2018
Last Updated
October 4, 2023
Sponsor
Astellas Pharma Global Development, Inc.
search

1. Study Identification

Unique Protocol Identification Number
NCT03505320
Brief Title
A Study to Assess the Antitumor Activity, Safety, Pharmacokinetics and Biomarkers of Zolbetuximab (IMAB362) in Participants With Claudin (CLDN) 18.2 Positive, Metastatic or Advanced Unresectable Gastric and Gastroesophageal Junction (GEJ) Adenocarcinoma
Acronym
ILUSTRO
Official Title
A Phase 2 Study of Zolbetuximab (IMAB362) as Monotherapy, in Combination With mFOLFOX6 (With or Without Nivolumab) and in Combination With Pembrolizumab in Subjects With Metastatic or Locally Advanced Unresectable Gastric or Gastroesophageal Junction (GEJ) Adenocarcinoma Whose Tumors Have High or Intermediate Claudin (CLDN) 18.2 Expression
Study Type
Interventional

2. Study Status

Record Verification Date
October 2023
Overall Recruitment Status
Recruiting
Study Start Date
June 29, 2018 (Actual)
Primary Completion Date
April 30, 2024 (Anticipated)
Study Completion Date
July 31, 2024 (Anticipated)

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
No

5. Study Description

Brief Summary
The purpose of this study is to determine the Objective Response Rate (ORR) of zolbetuzimab as a single agent as assessed by an independent central reader. This study will also assess the ORR and Progression Free Survival (PFS) of zolbetuximab in combination with mFOLFOX6 (with or without Nivolumab) and in combination with pembrolizumab, assess the safety and tolerability, assess the effects on CLDN18.2 expression and assess the immunogenicity and immunomodulatory effects of zolbetuximab as a single agent and in combination with mFOLFOX6 (with or without Nivolumab) and in combination with pembrolizumab. This study will also evaluate the pharmacokinetics (PK) of zolbetuximab, oxaliplatin, fluorouracil (5-FU), and pembrolizumab, evaluate health-Related Quality of Life (HRQoL), evaluate the Disease Control Rate (DCR), Duration of Response (DOR), PFS of zolbetuximab as a single agent, in combination with mFOLFOX6 (with or without Nivolumab) and in combination with pembrolizumab based on both investigator and independent central reader assessment, assess Overall Survival (OS) of zolbetuximab as a single agent and in combination with pembrolizumab.
Detailed Description
This is a study to assess the antitumor activity of zolbetuximab, an Immunoglobulin (IgG1) chimeric monoclonal antibody directed against CLDN18.2, in subjects with recurrent locally advanced or metastatic gastric or gastroesophageal junction adenocarcinoma whose tumors have high or intermediate CLDN18.2 expression. For each cohort, the study consists of the following periods: pre-screening; screening; treatment; and follow-up for disease progression. In addition, there will be a survival follow-up period for Cohort 1A and Cohort 4B participants only. Tolerability of zolbetuximab in combination with pembrolizumab in Japanese participant(s) will be evaluated in Cohort 3A DLT assessment. Tolerability of zolbetuximab in combination with mFOLFOX6 and nivolumab in Japanese subject(s) will be evaluated in Cohort 4B, if Japanese subjects are not enrolled in the Cohort 4A DLT assessment.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Pharmacokinetics of Zolbetuximab, Gastric Cancer, Gastro-esophageal Junction (GEJ) Cancer, Pharmacokinetics of Oxaliplatin, Pharmacokinetics of Fluorouracil Bolus (5-FU)
Keywords
zolbetuximab, oxaliplatin, fluorouracil, claudiximab, IMAB362, Gastro-Esophageal Junction cancer, Pembrolizumab, Gastric cancer

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 2
Interventional Study Model
Parallel Assignment
Masking
ParticipantInvestigator
Masking Description
Participants will be enrolled to receive zolbetuximab as monotherapy or in combination with mFOLFOX6 (with or without Nivolumab) and in combination with pembrolizumab in an unblinded fashion.
Allocation
Non-Randomized
Enrollment
116 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
zolbetuximab (Cohort 1A)
Arm Type
Experimental
Arm Description
Participants will be treated with zolbetuximab on a 21-day cycle in which zolbetuximab will be administered as a single agent every 3 weeks until disease progression, toxicity requiring cessation, start of another anti-cancer treatment or other treatment discontinuation criteria are met.
Arm Title
mFOLFOX6 plus zolbetuximab (Cohort 2)
Arm Type
Experimental
Arm Description
Participants will be treated with zolbetuximab and mFOLFOX6 on a 42-day cycle in which zolbetuximab is administered on days 1 and 22, and mFOLFOX6 is administered on days 1, 15 and 29; however, for the first cycle, zolbetuximab will be administered on day 3 (instead of day 1) to allow for pharmacokinetic collection. Participants will receive up to 12 mFOLFOX6 treatments (4 cycles). Beginning at cycle 5, participants may continue on 5-FU and leucovorin or folinic acid along with zolbetuximab for the remainder of the study per investigator's discretion. mFOLFOX6 treatment includes oxaliplatin: intravenous [IV] infusion, leucovorin: IV infusion, fluorouracil bolus: IV bolus, fluorouracil infusion: continuous IV infusion.
Arm Title
Pembrolizumab plus zolbetuximab (Cohort 3A)
Arm Type
Experimental
Arm Description
Participants will be treated with zolbetuximab and pembrolizumab on a 21-day cycle. Loading dose of zolbetuximab will be administered at cycle 1, day 1 followed by maintenance dose of zolbetuximab once every 3 weeks (Q3W). Pembrolizumab will be administered to 3 to 6 subjects at a intravenously on day 1 of every 21-day cycle and will be infused 1 hour after the zolbetuximab infusion is completed. Tolerability and safety of zolbetuximab in combination with pembrolizumab will be evaluated during the 3-week dose-limiting toxicity (DLT) assessment period. If this cycle 1 dose is not tolerable, a lower dose of zolbetuximab in combination with pembrolizumab will subsequently be evaluated.
Arm Title
Zolbetuximab in combination with mFOLFOX6 and nivolumab (Cohort 4A/4B)
Arm Type
Experimental
Arm Description
Participants will be treated with zolbetuximab and mFOLFOX6, nivolumab on a 42-day cycle. Cohort 4A: Loading dose of zolbetuximab in combination with nivolumab and mFOLFOX6 on cycle 1 day 1, followed by zolbetuximab in combination with nivolumab and mFOLFOX6 q2w [days 15 and 29] (1 cycle = 6 weeks). Tolerability and safety of zolbetuximab in combination with nivolumab, mFOLFOX6 will be evaluated during the 3-week DLT assessment period. If cycle 1 dose is not tolerable, a lower dose of dose zolbetuximab in combination with nivolumab and mFOLFOX6 will be subsequently evaluated. Cohort 4B: Subjects will be treated with the combination of zolbetuximab, mFOLFOX6 and nivolumab at the dose deemed tolerable in Cohort 4A. Subjects will receive up to 12 mFOLFOX6 treatments (4 cycles). For Cohorts 4A and 4B, beginning at cycle 5, subjects may continue on 5-FU and leucovorin or folinic acid along with zolbetuximab and nivolumab for the remainder of the study per investigator's discretion.
Intervention Type
Drug
Intervention Name(s)
zolbetuximab
Other Intervention Name(s)
IMAB362
Intervention Description
Zolbetuximab will be administered as a minimum 2-hour IV infusion.
Intervention Type
Drug
Intervention Name(s)
oxaliplatin
Intervention Description
Oxaliplatin will be administered as a 2-hour IV infusion.
Intervention Type
Drug
Intervention Name(s)
leucovorin
Intervention Description
Leucovorin will be administered as a 2-hour IV infusion.
Intervention Type
Drug
Intervention Name(s)
fluorouracil
Intervention Description
Fluorouracil will be administered as IV bolus over 5 to 15 minutes and continuous IV infusion over 46 to 48 hours or per institutional guidelines.
Intervention Type
Drug
Intervention Name(s)
Pembrolizumab
Intervention Description
Pembrolizumab will be administered intravenously over 30 minutes.
Intervention Type
Drug
Intervention Name(s)
folinic acid
Intervention Description
Folnic acid will be administered as a 2-hour IV infusion.
Intervention Type
Drug
Intervention Name(s)
nivolumab
Intervention Description
Nivolumab will be administered intravenously according to institutional standards.
Primary Outcome Measure Information:
Title
Objective Response Rate (ORR) of zolbetuximab as a single agent by central review (Cohort 1)
Description
The ORR is defined as the proportion of participants with complete or partial objective response based on Response Evaluation Criteria in Solid Tumors (RECIST) V1.1 (assessed by an independent review committee (IRC)).
Time Frame
Up to 3 months
Secondary Outcome Measure Information:
Title
Pharmacokinetics (PK) of zolbetuximab: Area Under the Concentration-time Curve from the Time of Dosing Extrapolated to Time Infinity (AUCinf) (Cohorts 1A, 2, 3A and 4)
Description
AUCinf will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Area Under the Concentration-time Curve from the Time of Dosing Extrapolated to Time Infinity Percentage (AUCinf (%extrap)) (Cohorts 1A, 2, 3A and 4)
Description
AUCinf (%extrap) will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Area Under the Concentration-time Curve from the Time of Dosing Until the Last Measurable Concentration (AUClast) (Cohorts 1A, 2, 3A and 4)
Description
AUClast will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Area Under the Concentration-Time Curve from the Time of Dosing to the Start of the Next Dosing Interval (AUCtau) (Cohorts 1A, 2, 3A and 4)
Description
AUCtau will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Maximum Concentration (Cmax) (Cohorts 1A, 2, 3A and 4)
Description
Cmax will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Concentration Immediately Prior to Dosing (Ctrough) (Cohorts 1A, 2, 3A and 4)
Description
Ctrough will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Time of Maximum Concentration (Tmax) (Cohorts 1A, 2, 3A and 4)
Description
Tmax will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Terminal Elimination Half-life (T1/2) (Cohorts 1A, 2, 3A and 4)
Description
T1/2 will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Time of the last measurable concentration (Tlast) (Cohorts 1A, 2, 3A and 4)
Description
Tlast will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Clearance (CL) (Cohorts 1A, 2, 3A and 4)
Description
CL will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of zolbetuximab: Volume of Distribution During the Terminal Phase (Vz) (Cohorts 1A, 2, 3A and 4)
Description
Vz will be derived from the PK serum samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: AUCinf (Cohort 2)
Description
AUCinf will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: AUCinf (%extrap) (Cohort 2)
Description
AUCinf (%extrap) will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: AUClast (Cohort 2)
Description
AUClast will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: Cmax (Cohort 2)
Description
Cmax will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: Tmax (Cohort 2)
Description
Tmax will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: T1/2 (Cohort 2)
Description
T1/2 will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: Tlast (Cohort 2)
Description
Tlast will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: CL (Cohort 2)
Description
TL will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of oxaliplatin: Vz (Cohort 2)
Description
Vz will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): AUCinf (Cohort 2)
Description
AUCinf will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): AUCinf (%extrap) (Cohort 2)
Description
AUCinf (%extrap) will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): AUClast (Cohort 2)
Description
AUClast will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): Cmax (Cohort 2)
Description
Cmax will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): Tmax (Cohort 2)
Description
Tmax will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): T1/2 (Cohort 2)
Description
T1/2 will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): Tlast (Cohort 2)
Description
Tlast will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): CL (Cohort 2)
Description
CL will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
PK of fluorouracil bolus (5-FU): Vz (Cohort 2)
Description
Vz will be derived from the PK plasma samples collected.
Time Frame
Up to 16 months
Title
Safety and tolerability assessed by adverse events (AEs) (Cohorts 1A, 2, 3A and 4)
Description
An AE is any untoward medical occurrence in a participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product.
Time Frame
Up to 16 months
Title
Number of participants with electrocardiogram (ECG) abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Description
Number of participants with potentially clinically significant ECG values.
Time Frame
Up to 14 months
Title
Number of participants with vital signs abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Description
Number of participants with potentially clinically significant vital sign values.
Time Frame
Up to 14 months
Title
Number of participants with European Cooperative Oncology Group (ECOG) performance status abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Description
Number of participants with potentially clinically significant ECOG performance status values. ECOG grades 0-5, where 0 = Fully active, able to carry on all pre-disease performance without restriction; 1 = Restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature, e.g., light house work, office work; 2 = Ambulatory and capable of all self-care but unable to carry out any work activities. Up and about more than 50% of waking hours; 3 = Capable of only limited self-care, confined to bed or chair more than 50% of waking hours; 4 = Completely disabled. Cannot carry on any self-care. Totally confined to bed or chair and 5 = Dead.
Time Frame
Up to 14 months
Title
Number of participants with laboratory assessments abnormalities and or adverse events (Cohorts 1A, 2, 3A and 4)
Description
Number of participants with potentially clinically significant laboratory values.
Time Frame
Up to 14 months
Title
Number of anti-drug antibody (ADA) Positive Participants (Cohorts 1A, 2, 3A and 4)
Description
Immunogenicity will be measured by the number of participants that are ADA positive.
Time Frame
Up to 16 months
Title
Health Related Quality of Life (HRQoL) measured by the Quality of Life Questionnaire - Core Questionnaire (QLQ-C30) questionnaire (Cohorts 1A, 2, 3A and 4)
Description
The EORTC-QLQ-C30 is a cancer-specific instrument consisting of 5 functional domain scales: physical, role, emotional, social and cognitive.
Time Frame
Up to 16 months
Title
HRQoL measured by the Oesophago-Gastric Module (EORTC QLQ-OG-25) questionnaire (Cohorts 1A, 2, 3A and 4)
Description
The EORTC-QLQ-OG25 instrument evaluates GC- and GEJC-specific symptoms such as stomach discomfort, difficulties eating and swallowing and indigestion.
Time Frame
Up to 16 months
Title
HRQoL measured by the Global Pain (GP) questionnaire (Cohorts 1A, 2, 3A and 4)
Description
The GP instrument is a single assessment of overall pain.
Time Frame
Up to 16 months
Title
HRQoL measured by the EuroQOL Five Dimensions Questionnaire 5L (EQ-5D-5L) questionnaire (Cohorts 1A, 2, 3A and 4)
Description
The EQ-5D-5L is a standardized instrument developed by the EuroQol Group for use as a generic, preference-based measure of health outcomes. The EQ-5D-5L is a 5-item self-reported measure of functioning and wellbeing, which assesses 5 dimensions of health, including mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension comprises 5 levels (no problems, slight problems, moderate problems, severe problems, extreme problems). A unique EQ-5D-5L health state is defined by combining 1 level from each of the 5 dimensions. This questionnaire also records the respondent's self-rated health status on a vertical graduated (0 = the worst health a participant can imagine to 100 = the best health a participant can imagine) visual analogue scale. Responses to the 5 items will also be converted to a weighted health state index (utility score) based on values derived from general population samples.
Time Frame
Up to 16 months
Title
HRQoL measured by the Health Resource Utilization (HRU) questionnaire (Cohorts 1A, 2, 3A and 4)
Description
Health resource utilization questionnaire to assess the number of office visits, hospital stays and other healthcare resource utilization that occur outside of the clinical trial.
Time Frame
Up to 16 months
Title
Disease Control Rate (DCR) of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
Description
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
Time Frame
Up to 3 months
Title
DCR of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
Description
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
Time Frame
Up to 13 months
Title
DCR of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
Description
DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
Time Frame
up to 13 months
Title
DCR of zolbetuximab as a single agent by independent central reader (Cohort 1A)
Description
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
Time Frame
Up to 3 months
Title
DCR of zolbetuximab in combination with mFOLFOX6 by Independent central reader (Cohort 2)
Description
The DCR is defined as the proportion of subjects with complete or partial objective response, or stable disease based on RECIST V1.1.
Time Frame
Up to 13 months
Title
Duration of Response (DOR) of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
Description
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
Time Frame
Up to 3 months
Title
DOR of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
Description
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
Time Frame
Up to 13 months
Title
DOR of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
Description
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
Time Frame
up to 13 months
Title
DOR of zolbetuximab as a single agent by independent central reader (Cohort 1A)
Description
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
Time Frame
Up to 3 months
Title
DOR of zolbetuximab in combination with mFOLFOX6 by independent central reader (Cohort 2)
Description
DOR is defined as the time from the date of the first response CR/PR (whichever is first recorded) to the date of radiographical progression/death or date of censoring.
Time Frame
Up to 13 months
Title
Progression Free Survival (PFS) of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
Description
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
Time Frame
Up to 3 months
Title
PFS of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
Description
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
Time Frame
Up to 13 months
Title
PFS of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
Description
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
Time Frame
up to 13 months
Title
PFS of zolbetuximab as a single agent by independent central review (Cohort 1A)
Description
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
Time Frame
Up to 3 months
Title
PFS of zolbetuximab in combination with mFOLFOX6 by independent central review (Cohort 2)
Description
PFS is defined as the time from date of treatment start until the date of radiological disease progression assessed by independent radiographic reviewer (IRR), or until death due to any cause.
Time Frame
Up to 13 months
Title
ORR of zolbetuximab as a single agent by investigator assessment (Cohort 1A)
Description
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
Time Frame
Up to 3 months
Title
ORR of zolbetuximab in combination with mFOLFOX6 by investigator assessment (Cohort 2)
Description
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
Time Frame
Up to 13 months
Title
ORR of zolbetuximab in combination with pembrolizumab by investigator assessment (Cohort 3A)
Description
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
Time Frame
Up to 5 months
Title
ORR of zolbetuximab in combination with mFOLFOX6 (with nivolumab) by investigator assessment (Cohort 4)
Description
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
Time Frame
up to 13 months
Title
ORR of zolbetuximab in combination with mFOLFOX6 by independent central reader (Cohort 2)
Description
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
Time Frame
Up to 13 months
Title
ORR of zolbetuximab in combination with pembrolizumab by independent central reader (Cohort 3A)
Description
The ORR is defined as the proportion of subjects with complete or partial objective response based on RECIST V1.1.
Time Frame
Up to 5 months
Title
Overall Survival (OS) of zolbetuximab as a single agent (Cohort 1A)
Description
OS is defined as the time from the date of treatment start until the documented date of death from any cause.
Time Frame
Up to 7 months
Title
OS of zolbetuximab in combination with mFOLFOX6 and nivolumab (Cohort 4B)
Description
OS is defined as the time from the date of treatment start until the documented date of death from any cause.
Time Frame
up to 56 Months

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Female subject eligible to participate if she is not pregnant and at least one of the following conditions applies: Not a woman of child-bearing potential (WOCBP) OR WOCBP who agrees to follow the contraceptive guidance throughout the treatment period and for at least 9 months after the final oxaliplatin administration and 6 months after the final administration of all other study drugs. 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 agree not to donate ova starting at screening and throughout the study period, and for 9 months after the final oxaliplatin administration and 6 months after the final administration of all other study drugs. A sexually active male subject with a female partner(s) who is of child-bearing potential must agree to use contraception during the treatment period and for at least 6 months after the final study drug administration. Male subject must agree not to donate sperm starting at screening and throughout the study period, and for 6 months after the final study drug administration. Male subject with a pregnant or breastfeeding partner(s) must agree to remain abstinent or use a condom for the duration of the pregnancy or time partner is breastfeeding throughout the study period and for 6 months after the final study drug administration. Subject has histologically confirmed gastric or GEJ adenocarcinoma. Subject has radiographically-confirmed, locally advanced, unresectable or metastatic disease within 28 days prior to the first dose of study treatment. Subject's tumor is positive for CLDN18.2 expression. Subject agrees to not participate in another interventional study while on treatment. Subject has ECOG performance status 0 to 1. Subject has predicted life expectancy ≥ 12 weeks. Subject must meet all of the following criteria based on the centrally or locally analyzed laboratory tests collected within 14 days prior to the first dose of study treatment. In case of multiple central laboratory data within this period, the most recent data should be used. Hemoglobin (Hgb) ≥ 9 g/dL (transfusion is allowed, but post-transfusion Hgb [24 hours or later following transfusion] must be ≥ 9 g/dL) Absolute neutrophil count (ANC) ≥ 1.5 × 109/L Platelets ≥ 100 × 10^9/L Albumin ≥ 2.5 g/dL Total bilirubin ≤ 1.5 × upper limit of normal (ULN) Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) ≤ 2.5 × ULN in subjects without liver metastases (≤ 5 × ULN if liver metastases are present) Estimated creatinine clearance ≥ 30 mL/min Prothrombin time/international normalized ratio and partial thromboplastin time ≤ 1.5 × ULN (except for subjects receiving anticoagulation therapy) Specific to Cohort 1A: Subject has measurable disease according to RECIST 1.1 within 28 days prior to the first dose of study treatment per investigator assessment. For subjects with only 1 evaluable lesion and prior radiotherapy ≤ 3 months before enrollment, the lesion must either be outside the field of prior radiotherapy or must have documented progression following radiation therapy. Subject has disease progression on or after at least 2 prior regimens for their advanced disease, including fluoropyrimidine and platinum-containing chemotherapy, and if appropriate, HER2/neu-targeted therapy and all associated side effects have resolved to grade 1 or less. Subject must have an additional available tumor specimen collected within 3 months prior to the first dose of study treatment. Subject must be an appropriate candidate for a tumor biopsy and is amenable to undergo a tumor biopsy during the screening period (if applicable) and treatment period as indicated in the Schedule of Assessments. Specific to Cohort 2: Subject has measurable disease according to RECIST 1.1 within 28 days prior to the first dose of study treatment per investigator assessment. For subjects with only 1 evaluable lesion and prior radiotherapy ≤ 3 months before enrollment, the lesion must either be outside the field of prior radiotherapy or must have documented progression following radiation therapy. Subject has not received prior systemic anti-cancer therapy for their advanced disease (subject may have received neoadjuvant and/or fluorouracil-containing adjuvant chemotherapy as long as it has been completed ≥ 6 months before the first dose of study treatment). Subject has a gastric or GEJ tumor that is HER2-negative as determined by local or central testing. Subject must have an additional available tumor specimen collected within 3 months prior to the first dose of study treatment. Subject must be an appropriate candidate for a tumor biopsy and is amenable to undergo a tumor biopsy during the screening period (if applicable) and treatment period as indicated in the Schedule of Assessments. Specific to Cohort 3A: Subject has radiologically evaluable disease (measurable and/or non-measurable) according to RECIST 1.1, per local assessment, ≤ 28 days prior to the first dose of study treatment. For subjects with only 1 evaluable lesion and prior radiotherapy ≤ 3 months before enrollment, the lesion must either be outside the field of prior radiotherapy or must have documented progression following radiation therapy. Subject has disease progression on or after at least 2 prior regimens for their advanced disease, including fluoropyrimidine and platinum-containing chemotherapy, and if appropriate, HER2/neu-targeted therapy. Subject has not received prior checkpoint inhibitor therapy. Specific to Cohort 4A and 4B: Subject has radiologically evaluable disease. Subject has not received prior systemic anti-cancer therapy for their advanced disease. Subject has a gastric or GEJ tumor that is HER2-negative as determined by local or central testing. Subject has not received prior checkpoint inhibitor therapy. Specific to Cohort 4B Only: Subject must have an additional available tumor specimen collected within 3 months prior to the first dose of study treatment. Subject must be an appropriate candidate for a tumor biopsy and is amenable to undergo a tumor biopsy during the screening period (if applicable) and treatment period. Subject has a tumor that is PD-L1 positive. Exclusion Criteria: Subject has had prior severe allergic reaction or intolerance to known ingredients of zolbetuximab or other monoclonal antibodies, including humanized or chimeric antibodies. Subject has known immediate or delayed hypersensitivity or contraindication to any component of study treatment. Subject has received other investigational agents or devices concurrently or within 28 days prior to first dose of study treatment. Subject has received systemic immunosuppressive therapy, including systemic corticosteroids 14 days prior to first dose of study treatment. Subject has a complete gastric outlet syndrome or a partial gastric outlet syndrome with persistent recurrent vomiting. Subject has significant gastric bleeding and/or untreated gastric ulcers that would preclude the subject from participation. Subject has history of central nervous system metastases and/or carcinomatous meningitis from gastric/GEJ cancer. Subject has a known history of a positive test for human immunodeficiency virus (HIV) infection or known active hepatitis B (positive hepatitis B surface antigen [HBsAg]) or hepatitis C infection. Subject has had within 6 months prior to first dose of study treatment any of the following: unstable angina, myocardial infarction, ventricular arrhythmia requiring intervention or hospitalization for heart failure. Subject has active infection requiring systemic therapy that has not completely resolved within 7 days prior to the start of study treatment. Subject has active autoimmune disease that has required systemic treatment within the past 3 months prior to the start of study treatment. Subject has a clinically significant disease or co-morbidity that may adversely affect the safe delivery of treatment within this study or make the subject unsuitable for study participation. Subject has psychiatric illness or social situations that would preclude study compliance. Subject has had a major surgical procedure ≤ 28 days before start of study treatment. Subject is without complete recovery from a major surgical procedure ≤ 14 days before start of study treatment Subject has received radiotherapy for locally advanced unresectable or metastatic gastric or GEJ adenocarcinoma ≤ 14 days (Cohorts 1 and 3A) and ≤ 28 days (Cohorts 2 and 4A or 4B) prior to start of study treatment and has NOT recovered from any related toxicity. Subject has another malignancy, for which treatment is required. Cohort 2 and 4 Only, subject has any of the following: Prior severe allergic reaction or intolerance to any component of mFOLFOX6 chemotherapeutics in this study Known dihydropyrimidine dehydrogenase deficiency (DPD). Known peripheral neuropathy > Grade 1 (absence of deep tendon reflexes as the sole neurological abnormality does not render the subject ineligible). Sinusoidal obstruction syndrome, formerly known as veno-occlusive disease, if present, should be stable or improving. History of clinically significant ventricular arrhythmias. QTc interval > 450 msec for male subjects; QTc interval > 470 msec for female subjects. History or family history of congenital long QT syndrome. Cardiac arrhythmias requiring anti-arrhythmic medications (Subjects with rate controlled atrial fibrillation for > 1 month prior to first dose of study treatment are eligible). Cohorts 3A, 4A and 4B Only, subject has any of the following: Subjects with ongoing or previous autoimmune disease or interstitial lung disease, active diverticulitis or gastrointestinal ulcerative disease, or solid organ or stem cell transplant (for Cohort 4) or other uncontrolled or clinically significant medical disorders. Subjects with type 1 diabetes mellitus, endocrinopathies stably maintained on appropriate replacement therapy or skin disorders (e.g., vitiligo, psoriasis, or alopecia) not requiring systemic treatment are allowed. Subject has known history of serious hypersensitivity reaction to a known ingredient of pembrolizumab or nivolumab. Cohort 4B Only: Subject with known microsatellite instability-high or mismatch repair deficient tumors.
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Astellas Pharma Global Development
Phone
800-888-7704
Email
astellas.registration@astellas.com
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Global Medical Lead
Organizational Affiliation
Astellas Pharma Global Development, Inc.
Official's Role
Study Director
Facility Information:
Facility Name
The Angeles Clinic and Research Institute
City
Los Angeles
State/Province
California
ZIP/Postal Code
90025
Country
United States
Individual Site Status
Withdrawn
Facility Name
UCLA Medical Center
City
Santa Monica
State/Province
California
ZIP/Postal Code
90404
Country
United States
Individual Site Status
Withdrawn
Facility Name
Georgetown Univ Hospital
City
Washington
State/Province
District of Columbia
ZIP/Postal Code
20007
Country
United States
Individual Site Status
Withdrawn
Facility Name
University of Chicago
City
Chicago
State/Province
Illinois
ZIP/Postal Code
60637
Country
United States
Individual Site Status
Recruiting
Facility Name
Indiana University Cancer Center
City
Indianapolis
State/Province
Indiana
ZIP/Postal Code
46077
Country
United States
Individual Site Status
Withdrawn
Facility Name
Mass General / North Shore Can
City
Boston
State/Province
Massachusetts
ZIP/Postal Code
02114
Country
United States
Individual Site Status
Recruiting
Facility Name
Karmanos Cancer Institute
City
Detroit
State/Province
Michigan
ZIP/Postal Code
48201
Country
United States
Individual Site Status
Withdrawn
Facility Name
Memorial Sloan Kettering Cancer Center
City
New York
State/Province
New York
ZIP/Postal Code
10065
Country
United States
Individual Site Status
Withdrawn
Facility Name
Weill Cornell Medical College
City
New York
State/Province
New York
ZIP/Postal Code
10065
Country
United States
Individual Site Status
Suspended
Facility Name
Sanford Cancer Center
City
Sioux Falls
State/Province
South Dakota
ZIP/Postal Code
57104
Country
United States
Individual Site Status
Withdrawn
Facility Name
Virginia Cancer Specialists
City
Fairfax
State/Province
Virginia
ZIP/Postal Code
22031
Country
United States
Individual Site Status
Suspended
Facility Name
Site FR33003
City
Pessac
State/Province
Nouvelle-Aquitaine
ZIP/Postal Code
33604
Country
France
Individual Site Status
Recruiting
Facility Name
Site FR33002
City
Poitiers
State/Province
Nouvelle-Aquitaine
ZIP/Postal Code
86021
Country
France
Individual Site Status
Recruiting
Facility Name
Site FR33001
City
Brest
Country
France
Individual Site Status
Recruiting
Facility Name
Site FR33004
City
Paris
ZIP/Postal Code
75015
Country
France
Individual Site Status
Recruiting
Facility Name
Site IT39005
City
Milano
Country
Italy
Individual Site Status
Recruiting
Facility Name
Site IT39002
City
Napoli
Country
Italy
Individual Site Status
Suspended
Facility Name
Site IT39004
City
Padova
Country
Italy
Individual Site Status
Suspended
Facility Name
Site IT39003
City
Pisa
Country
Italy
Individual Site Status
Suspended
Facility Name
Site JP81001
City
Chiba
Country
Japan
Individual Site Status
Recruiting
Facility Name
Site JP81002
City
Tokyo
Country
Japan
Individual Site Status
Recruiting
Facility Name
Site JP81003
City
Tokyo
Country
Japan
Individual Site Status
Recruiting
Facility Name
Site KR82002
City
Seongnam-si
Country
Korea, Republic of
Individual Site Status
Recruiting
Facility Name
Site KR82001
City
Seoul
Country
Korea, Republic of
Individual Site Status
Recruiting
Facility Name
Site TW88601
City
Taichung
Country
Taiwan
Individual Site Status
Recruiting
Facility Name
Site TW88602
City
Tainan
ZIP/Postal Code
70403
Country
Taiwan
Individual Site Status
Suspended

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/

Learn more about this trial

A Study to Assess the Antitumor Activity, Safety, Pharmacokinetics and Biomarkers of Zolbetuximab (IMAB362) in Participants With Claudin (CLDN) 18.2 Positive, Metastatic or Advanced Unresectable Gastric and Gastroesophageal Junction (GEJ) Adenocarcinoma

We'll reach out to this number within 24 hrs