MedPath

Tucatinib, Trastuzumab, Ramucirumab, and Paclitaxel Versus Paclitaxel and Ramucirumab in Previously Treated HER2+ Gastroesophageal Cancer

Phase 2
Completed
Conditions
Gastric Adenocarcinoma
Gastroesophageal Junction Adenocarcinoma
Esophageal Adenocarcinoma
Interventions
Other: tucatinib placebo
Other: trastuzumab placebo
Registration Number
NCT04499924
Lead Sponsor
Seagen Inc.
Brief Summary

This study is being done to see if tucatinib with trastuzumab, ramucirumab and paclitaxel works better than ramucirumab and paclitaxel to treat HER2-positive (HER2+) cancer of the gut (stomach or gastroesophageal cancer). This study will also look at what side effects happen when participants take this combination of drugs. A side effect is anything the drug does other than treating cancer.

Study treatment will be given in 28-day cycles.

In the Phase 2 part of the trial, participants and their doctors will know what drugs are being given (open-label). In the Phase 3 part, the study is "blinded." This means that participants, their doctor, and the study sponsor will not know which drugs are being given.

Detailed Description

Not available

Recruitment & Eligibility

Status
COMPLETED
Sex
All
Target Recruitment
17
Inclusion Criteria
  • Histologically or cytologically confirmed diagnosis of locally-advanced unresectable or metastatic HER2+ gastric or gastroesophageal junction adenocarcinoma (GEC)

  • HER2+ disease documented since progression of the most recent line of systemic therapy, as follows:

    • Phase 2 paclitaxel dose optimization stage:

      • HER2 amplification in a blood-based NGS assay performed at a central laboratory, or
      • HER2 overexpression/amplification immunohistochemistry (IHC) and in situ hybridization (ISH) (IHC3+ or IHC2+/ISH+) assay of a tumor tissue sample
    • Phase 2 dose expansion stage:

      • Cohort 2A: HER2 amplification in a blood-based NGS assay performed at a central laboratory
      • Cohort 2B: No HER2 amplification by blood-based NGS assay, but HER2 overexpression/amplification by IHC and ISH (IHC3+ or IHC2+/ISH+) assay of a tumor tissue sample
    • Phase 3: HER2 amplification in a blood-based NGS assay performed at a central laboratory

  • History of prior treatment with a HER2-directed antibody

  • Progressive disease during or after first-line therapy for locally-advanced unresectable or metastatic GEC

  • Phase 2: Measurable disease according to RECIST version 1.1

  • Phase 3: Measurable or non-measurable disease according to RECIST version 1.1

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

  • Life expectancy of at least 3 months, in the opinion of the investigator

Exclusion Criteria
  • Subjects with squamous cell or undifferentiated GEC
  • Having received more than 1 line of prior systemic therapy for locally-advanced unresectable or metastatic disease
  • Having received taxanes ≤12 months prior to enrollment, prior treatment with ramucirumab, or prior treatment with tucatinib, lapatinib, neratinib, afatinib, or any other investigational anti-HER2 and/or anti-EGFR tyrosine kinase inhibitor, or with T-DM1, T-Dxd, or any other HER2-directed antibody-drug conjugate
  • Phase 2 paclitaxel dose optimization stage only: history of prior partial or total gastrectomy
  • Unable to swallow pills

Study & Design

Study Type
INTERVENTIONAL
Study Design
PARALLEL
Arm && Interventions
GroupInterventionDescription
Phase 2 ArmtrastuzumabTucatinib + trastuzumab + ramucirumab + paclitaxel
Phase 2 ArmramucirumabTucatinib + trastuzumab + ramucirumab + paclitaxel
Phase 2 ArmpaclitaxelTucatinib + trastuzumab + ramucirumab + paclitaxel
Arm 3AtrastuzumabTucatinib + trastuzumab + ramucirumab + paclitaxel
Arm 3Btrastuzumab placeboRamucirumab + paclitaxel + tucatinib placebo + trastuzumab placebo
Arm 3CtucatinibTucatinib + ramucirumab + paclitaxel + trastuzumab placebo
Arm 3Btucatinib placeboRamucirumab + paclitaxel + tucatinib placebo + trastuzumab placebo
Arm 3CpaclitaxelTucatinib + ramucirumab + paclitaxel + trastuzumab placebo
Arm 3BramucirumabRamucirumab + paclitaxel + tucatinib placebo + trastuzumab placebo
Arm 3BpaclitaxelRamucirumab + paclitaxel + tucatinib placebo + trastuzumab placebo
Arm 3CramucirumabTucatinib + ramucirumab + paclitaxel + trastuzumab placebo
Arm 3Ctrastuzumab placeboTucatinib + ramucirumab + paclitaxel + trastuzumab placebo
Phase 2 ArmtucatinibTucatinib + trastuzumab + ramucirumab + paclitaxel
Arm 3AtucatinibTucatinib + trastuzumab + ramucirumab + paclitaxel
Arm 3AramucirumabTucatinib + trastuzumab + ramucirumab + paclitaxel
Arm 3ApaclitaxelTucatinib + trastuzumab + ramucirumab + paclitaxel
Primary Outcome Measures
NameTimeMethod
Number of Participants With Dose-limiting Toxicities (DLT) During the First Cycle of TreatmentCycle 1 (28 days)

DLTs were adverse events (AEs), laboratory abnormalities, or treatment modifications that occurred during the first cycle of treatment in the Phase 2 paclitaxel dose optimization stage that were related to paclitaxel, to tucatinib, or to the combination of tucatinib, trastuzumab, ramucirumab and paclitaxel and that met any of the study protocol specified criteria. The relationship of AEs to study drugs was determined by the investigator. AEs that were attributed only to trastuzumab and/or ramucirumab, but not tucatinib or paclitaxel were not considered DLTs.

Number of Participants With Treatment-emergent Adverse Events (TEAEs)From first dose of the study treatment (Day 1) up to 30 days after the last dose of study treatment (maximum treatment duration= 19.8 months, maximum follow-up duration up to 20.8 months)

An AE was any untoward medical occurrence in a participant or clinical investigational participant administered a medicinal product and which did not necessarily have a causal relationship with this treatment. AEs were defined as treatment emergent if they were newly occurring or worsened following study treatment. A TEAE was defined as a newly occurring or worsening AE after the first dose of study treatment (tucatinib, trastuzumab, ramucirumab, or paclitaxel) and up to 30 days after the last dose of study treatment (tucatinib, trastuzumab, ramucirumab, or paclitaxel, whichever was later).

Maximum Percentage Change From Baseline in WeightFrom Baseline (Day 1) up to 30 days after the last dose of study treatment (maximum treatment duration= 19.8 months, maximum follow-up duration up to 20.8 months)

Maximum percentage decrease from baseline in weight is reported in this outcome measure.

Number of Participants With Any Dose ModificationsFrom first dose of the study treatment (Day 1) up to the last dose of study treatment (maximum treatment duration up to 19.8 months)

Dose modification included dose hold, dose reduction, dose error and unplanned dose adjustments. Number of participants with any dose treatment modifications for paclitaxel, ramucirumab, trastuzumab, and tucatinib are reported in this outcome measure.

Number of Participants With Treatment-emergent Laboratory AbnormalitiesFrom first dose of the study treatment (Day 1) up to 30 days after the last dose of study treatment (maximum treatment duration= 19.8 months, maximum follow-up duration up to 20.8 months)

Treatment-emergent laboratory abnormalities were defined as abnormalities that are new or worsened on or after receiving the first dose of study treatment up to 30 days after the last dose of study treatment. Laboratory abnormalities included: 1) Blood chemistry: alanine aminotransferase increased, albumin decreased, alkaline phosphatase increased, aspartate aminotransferase increased, calcium corrected for albumin decreased, calcium corrected for albumin increased, creatinine increased, glomerular filtration rate estimated decreased, glucose decreased, lactate dehydrogenase increased, magnesium decreased, magnesium increased, potassium decreased, potassium increased, sodium decreased, sodium increased, and total bilirubin increased; 2) Hematological: hemoglobin decreased, leukocytes decreased, lymphocytes decreased, neutrophils decreased, and platelets decreased.

Number of Participants With Clinically Significant Vital Signs ValuesFrom first dose of the study treatment (Day 1) up to 30 days after the last dose of study treatment (maximum treatment duration= 19.8 months, maximum follow-up duration up to 20.8 months)

Vital sign examinations included systolic blood pressure (SBP), diastolic blood pressure (DBP), heart rate. Criteria: SBP greater than or equal to (\>=) 120 millimeters of mercury (mm Hg) or DBP \>= 80 mm Hg; SBP \>= 140 mm Hg or DBP \>= 90 mm Hg; SBP \>= 160 mm Hg or DBP \>= 100 mm Hg), heart rate greater than (\>) 100 beats per minute (min). Clinical significance in vital signs abnormalities was judged by investigator. In this outcome measure number of participants with at least 1 clinically significant abnormality in any vital sign are reported.

Secondary Outcome Measures
NameTimeMethod
Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v) 1.1 By Investigator AssessmentFrom the first dose of study treatment until the first documented CR or PR on or before the first documented PD or new anti-cancer therapies or death, whichever occurred first (up to 19.8 months)

ORR was defined as the percentage of participants with best overall response of complete response (CR) or partial response (PR) according to RECIST v 1.1 by investigator assessment. As per RECIST v1.1, CR was defined as disappearance of all target and non-target lesions and normalization of tumor markers. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to less than (\<) 10 millimeter (mm). PR was defined as at least a 30 % decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. As per RECIST v1.1, disease progression (PD) was defined as one of the following: at least a 20% increase in the sum of diameters of target lesions (taking as reference the smallest sum on study) with an absolute increase of at least 5mm, unequivocal progression of existing non-target lesions, or the appearance of one or more new lesions.

Confirmed ORR Per RECIST v1.1 By Investigator AssessmentFrom the first dose of study treatment until the first documented confirmed CR or PR or before the first documented PD or new anti-cancer therapies or death, whichever occurred first (up to 19.8 months)

Confirmed ORR was defined as the percentage of participants with best overall response of confirmed CR or PR according to RECIST v1.1 by investigator assessment. For a response to be confirmed, the subsequent response needs to be at least 4 weeks after the initial response. As per RECIST v1.1, CR was defined as disappearance of all target and non-target lesions and normalization of tumor markers. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to \< 10 mm. PR was defined as at least a 30 % decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. As per RECIST v1.1, disease progression (PD) was defined as one of the following: at least a 20% increase in the sum of diameters of target lesions (taking as reference the smallest sum on study) with an absolute increase of at least 5mm, unequivocal progression of existing non-target lesions, or the appearance of one or more new lesions.

Progression-Free Survival (PFS) Per RECIST v1.1 By Investigator AssessmentFrom the date of first dose until the first documentation of PD or death or censoring date, whichever occurred first (up to 19.8 months)

PFS was defined as the time from the date of treatment initiation to the date of documented PD or death from any cause, whichever occurred first per RECIST version 1.1 by investigator assessment. As per RECIST v1.1, disease progression (PD) was defined as one of the following: at least a 20% increase in the sum of diameters (SOD) of target lesions (taking as reference the smallest sum on study) with an absolute increase of at least 5mm, unequivocal progression of existing non-target lesions, or the appearance of one or more new lesions. Participants without documentation of PD or death at the time of analysis were censored at the date of the last tumor assessment. Kaplan-Meier method was used for evaluation.

Duration of Response (DOR) Per RECIST v1.1 By Investigator AssessmentFrom the first documented CR or PR until the first documentation of PD or death or censoring date, whichever occurred first (up to 19.8 months)

DOR: time from first documentation of objective response of CR or PR to first documentation of PD or death from any cause, whichever occurred first according to RECIST v1.1 by investigator assessment. As per RECIST v1.1, CR: disappearance of all target and non-target lesions and normalization of tumor markers. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to \<10mm. PR: at least a 30% decrease in sum of diameters of target lesions, taking as reference baseline sum diameters. PD was defined as one of the following: at least a 20% increase in sum of diameters of target lesions (taking as reference the smallest sum on study) with an absolute increase of at least 5mm, unequivocal progression of existing non-target lesions, or appearance of one or more new lesions. Participants without documentation of PD or death at time of analysis were censored at the date of last tumor assessment. Kaplan-Meier method was used for evaluation.

Disease Control Rate (DCR) Per RECIST v1.1 By Investigator AssessmentFrom the first dose study treatment until PD or death, whichever occurred first (up to 19.8 months)

DCR was defined as percentage of participants with CR, PR, or stable disease (SD or non-CR/non-progressive disease) according to RECIST v1.1 by investigator assessment. As per RECIST v1.1, CR was defined as disappearance of all target and non-target lesions and normalization of tumor markers. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to \< 10 mm. PR was defined as at least a 30 % decrease in sum of diameters of target lesions, taking as reference the baseline sum diameters. PD was defined as one of following: at least a 20% increase in the sum of diameters of target lesions (taking as reference the smallest sum on study) with an absolute increase of at least 5mm, unequivocal progression of existing non-target lesions, or appearance of one or more new lesions. SD was defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference smallest sum of diameters.

Area Under the Plasma Concentration-time Curve to the Time of the Last Quantifiable Concentration (AUClast) of Paclitaxel, Tucatinib and Their MetabolitesTucatinib and ONT-993- Cycle 1 Day 8, Cycle 2 Day 1: Pre-dose, 2, 4, 6, 8 hours post-dose; Paclitaxel and its metabolites- Cycle 1 Days 1 and 8, Cycle 2 Day 1: Pre-dose, 2, 4, 6, 8 hours post-dose (each cycle length=28 days)

AUClast was reported for tucatinib, tucatinib metabolite (ONT-993), paclitaxel and paclitaxel metabolites (6 alpha-hydroxy-paclitaxel, 3'-p-hydroxy-paclitaxel and 6 alpha-3'-p-Dihydroxy-paclitaxel).

Maximum Observed Plasma Concentration (Cmax)Tucatinib and ONT-993- Cycle 1 Day 8, Cycle 2 Day 1: Pre-dose, 2, 4, 6, 8 hours post-dose (each cycle length=28 days)

Cmax was reported for tucatinib and tucatinib metabolite ONT-993.

Time to Maximum Observed Plasma Concentration (Tmax)Tucatinib and ONT-993- Cycle 1 Day 8, Cycle 2 Day 1: Pre-dose, 2, 4, 6, 8 hours post-dose (each cycle length=28 days)

Tmax was reported for tucatinib and tucatinib metabolite ONT-993.

Trough Concentration (Ctrough)Tucatinib and ONT-993- Cycle 1 Day 8, Cycle 2 Day 1: Predose (each cycle length=28 days)

Ctrough was calculated for tucatinib and tucatinib metabolite ONT-993.

Metabolite Ratio Based on AUClast (MRAUClast)Paclitaxel and its metabolites- Cycle 1 Days 1 and 8, Cycle 2 Day 1: Pre-dose, 2, 4, 6, 8 hours post-dose (each cycle length=28 days)

MRAUClast was defined as metabolic ratio, that is, ratio of the AUClast of a given paclitaxel metabolite over the AUClast of paclitaxel. MRAUClast for 6 alpha-hydroxypaclitaxel, 3-p-hydroxy-paclitaxel and 6 alpha-3-p-dihydroxy-paclitaxel was reported in this outcome measure.

Trial Locations

Locations (48)

University of Chicago Medical Center

🇺🇸

Chicago, Illinois, United States

Comprehensive Cancer Centers of Nevada

🇺🇸

Las Vegas, Nevada, United States

Duke University Medical Center

🇺🇸

Durham, North Carolina, United States

Cancer Centers of Colorado - Denver

🇺🇸

Denver, Colorado, United States

Lutheran Medical Center - Cancer Centers of Colorado

🇺🇸

Wheat Ridge, Colorado, United States

SCL Health - St. Mary's Hospital & Medical Center

🇺🇸

Grand Junction, Colorado, United States

Rocky Mountain Cancer Centers - Aurora

🇺🇸

Aurora, Colorado, United States

SCL Health Good Samaritan Medical Center Cancer Centers of Colorado

🇺🇸

Lafayette, Colorado, United States

Mayo Clinic Arizona

🇺🇸

Phoenix, Arizona, United States

McGill University Department of Oncology / McGill University Health Centre

🇨🇦

Montreal, Quebec, Canada

Dong-A University Hospital

🇰🇷

Busan, Korea, Republic of

Oncology Associates of Oregon

🇺🇸

Eugene, Oregon, United States

Centre Hospitalier de l'Universite de Montreal

🇨🇦

Montreal, Quebec, Canada

Lombardi Cancer Center / Georgetown University Medical Center

🇺🇸

Washington, District of Columbia, United States

Kyungpook National University Chilgok Hospital

🇰🇷

Daegu, Korea, Republic of

Dana Farber Cancer Institute

🇺🇸

Boston, Massachusetts, United States

Severance Hospital, Yonsei University Health System

🇰🇷

Seoul, Korea, Republic of

University of Tennessee

🇺🇸

Knoxville, Tennessee, United States

City of Hope National Medical Center

🇺🇸

Duarte, California, United States

Seoul National University Bundang Hospital

🇰🇷

Seongnam-si, Korea, Republic of

Asan Medical Center - Oncology

🇰🇷

Seoul, Korea, Republic of

University of Alabama at Birmingham

🇺🇸

Birmingham, Alabama, United States

Minnesota Oncology Hematology P.A.

🇺🇸

Saint Paul, Minnesota, United States

Allegheny General Hospital

🇺🇸

Pittsburgh, Pennsylvania, United States

Tennessee Oncology-Nashville/Sarah Cannon Research Institute

🇺🇸

Nashville, Tennessee, United States

Texas Oncology - Baylor Sammons Cancer Center

🇺🇸

Dallas, Texas, United States

Texas Oncology - San Antonio Medical Center

🇺🇸

San Antonio, Texas, United States

Huntsman Cancer Institute/University of Utah

🇺🇸

Salt Lake City, Utah, United States

Texas Oncology - Tyler

🇺🇸

Tyler, Texas, United States

Northwest Cancer Specialists, P.C.

🇺🇸

Vancouver, Washington, United States

London Regional Cancer Program, London Health Sciences Centre

🇨🇦

London, Ontario, Canada

Central Coast Local Health District (Gosford and Wyong Hospitals)

🇦🇺

Gosford, Australia

Austin Health

🇦🇺

Heidelberg, Australia

Seoul National University Hospital

🇰🇷

Seoul, Korea, Republic of

Samsung Medical Center

🇰🇷

Seoul, Korea, Republic of

Ajou University Hospital

🇰🇷

Suwon-si, Korea, Republic of

National Taiwan University Hospital

🇨🇳

Taipei, Taiwan

Kaohsiung Medical University Hospital

🇨🇳

Kaohsiung, Taiwan

Taipei Veterans General Hospital

🇨🇳

Taipei, Taiwan

The Christie NHS Foundation Trust

🇬🇧

Manchester, United Kingdom

Arizona Cancer Center / University of Arizona

🇺🇸

Tucson, Arizona, United States

UCLA Medical Center / David Geffen School of Medicine

🇺🇸

Santa Monica, California, United States

Norton Cancer Institute

🇺🇸

Louisville, Kentucky, United States

Holden Comprehensive Cancer Center / University of Iowa

🇺🇸

Iowa City, Iowa, United States

Roswell Park Cancer Institute

🇺🇸

Buffalo, New York, United States

Cleveland Clinic - Taussig Cancer Institute

🇺🇸

Cleveland, Ohio, United States

University of Pennsylvania / Perelman Center for Advanced Medicine

🇺🇸

Philadelphia, Pennsylvania, United States

Sarah Cannon Research Institute UK

🇬🇧

London, United Kingdom

© Copyright 2025. All Rights Reserved by MedPath