Clinical Trials Logo

Clinical Trial Details — Status: Active, not recruiting

Administrative data

NCT number NCT04499924
Other study ID # SGNTUC-022
Secondary ID
Status Active, not recruiting
Phase Phase 2/Phase 3
First received
Last updated
Start date March 22, 2021
Est. completion date March 31, 2027

Study information

Verified date August 2023
Source Seagen Inc.
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial 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.


Recruitment information / eligibility

Status Active, not recruiting
Enrollment 17
Est. completion date March 31, 2027
Est. primary completion date May 31, 2025
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility 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


Related Conditions & MeSH terms


Intervention

Drug:
tucatinib
300 mg given twice daily orally
trastuzumab
6 mg/kg loading dose will be administered intravenously (IV; into the vein) on Cycle 1 Day 1, followed by 4 mg/kg IV on Cycle 1 Day 15 and then Days 1 and 15 of each cycle thereafter
ramucirumab
8 mg/kg will be administered IV on Days 1 and 15 of each cycle
paclitaxel
60 or 80 mg/m^2 IV on Days 1, 8, and 15 of each cycle
Other:
tucatinib placebo
Given twice daily orally
trastuzumab placebo
IV on Days 1 and 15 of each cycle

Locations

Country Name City State
Australia Central Coast Local Health District (Gosford and Wyong Hospitals) Gosford
Australia Austin Health Heidelberg
Canada London Regional Cancer Program, London Health Sciences Centre London Ontario
Canada Centre Hospitalier de l'Universite de Montreal Montreal Quebec
Canada McGill University Department of Oncology / McGill University Health Centre Montreal Quebec
Korea, Republic of Dong-A University Hospital Busan
Korea, Republic of Kyungpook National University Chilgok Hospital Daegu
Korea, Republic of Seoul National University Bundang Hospital Seongnam-si
Korea, Republic of Asan Medical Center - Oncology Seoul
Korea, Republic of Samsung Medical Center Seoul
Korea, Republic of Seoul National University Hospital Seoul
Korea, Republic of Severance Hospital, Yonsei University Health System Seoul
Korea, Republic of Ajou University Hospital Suwon-si
Taiwan Kaohsiung Medical University Hospital Kaohsiung
Taiwan National Taiwan University Hospital Taipei
Taiwan Taipei Veterans General Hospital Taipei
United Kingdom Sarah Cannon Research Institute UK London
United Kingdom The Christie NHS Foundation Trust Manchester
United States Rocky Mountain Cancer Centers - Aurora Aurora Colorado
United States University of Alabama at Birmingham Birmingham Alabama
United States Dana Farber Cancer Institute Boston Massachusetts
United States Roswell Park Cancer Institute Buffalo New York
United States University of Chicago Medical Center Chicago Illinois
United States Cleveland Clinic - Taussig Cancer Institute Cleveland Ohio
United States Texas Oncology - Baylor Sammons Cancer Center Dallas Texas
United States Cancer Centers of Colorado - Denver Denver Colorado
United States City of Hope National Medical Center Duarte California
United States Duke University Medical Center Durham North Carolina
United States Oncology Associates of Oregon Eugene Oregon
United States SCL Health - St. Mary's Hospital & Medical Center Grand Junction Colorado
United States Holden Comprehensive Cancer Center / University of Iowa Iowa City Iowa
United States University of Tennessee Knoxville Tennessee
United States SCL Health Good Samaritan Medical Center Cancer Centers of Colorado Lafayette Colorado
United States Comprehensive Cancer Centers of Nevada Las Vegas Nevada
United States Norton Cancer Institute Louisville Kentucky
United States Tennessee Oncology-Nashville/Sarah Cannon Research Institute Nashville Tennessee
United States University of Pennsylvania / Perelman Center for Advanced Medicine Philadelphia Pennsylvania
United States Mayo Clinic Arizona Phoenix Arizona
United States Allegheny General Hospital Pittsburgh Pennsylvania
United States Minnesota Oncology Hematology P.A. Saint Paul Minnesota
United States Huntsman Cancer Institute/University of Utah Salt Lake City Utah
United States Texas Oncology - San Antonio Medical Center San Antonio Texas
United States UCLA Medical Center / David Geffen School of Medicine Santa Monica California
United States Arizona Cancer Center / University of Arizona Tucson Arizona
United States Texas Oncology - Tyler Tyler Texas
United States Northwest Cancer Specialists, P.C. Vancouver Washington
United States Lombardi Cancer Center / Georgetown University Medical Center Washington District of Columbia
United States Lutheran Medical Center - Cancer Centers of Colorado Wheat Ridge Colorado

Sponsors (1)

Lead Sponsor Collaborator
Seagen Inc.

Countries where clinical trial is conducted

United States,  Australia,  Canada,  Korea, Republic of,  Taiwan,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Overall survival (OS) (Phase 3 only) OS is defined as the time from randomization to death due to any cause 60 months
Primary Progression-free survival (PFS) per Response evaluation criteria in solid tumors (RECIST) version 1.1 according to investigator assessment (Phase 3 only) PFS is defined as the time from randomization to the date of disease progression or death from any cause 36 months
Primary Incidence of dose-limiting toxicities (DLTs) (Phase 2 only) During first cycle of treatment; up to one month
Primary Incidence of adverse events (AEs) (Phase 2 only) An adverse event is any untoward medical occurrence in a subject or clinical investigational subject administered a medicinal product and which does not necessarily have a causal relationship with this treatment. 18 months
Primary Incidence of laboratory abnormalities (Phase 2 only) To be summarized using descriptive statistics. 18 months
Primary Incidence of dose modifications (Phase 2 only) 18 months
Secondary Confirmed objective response rate (ORR) per RECIST version 1.1 according to investigator assessment (Phases 2 and 3) ORR is defined as the proportion of subjects with best overall response of CR or PR 36 months
Secondary ORR per RECIST version 1.1 according to investigator assessment (Phases 2 and 3) ORR is defined as the proportion of subjects with best overall response of CR or PR 36 months
Secondary Duration of response (DOR) per RECIST version 1.1 according to investigator assessment (Phases 2 and 3) DOR is defined as the time from first documentation of objective response of CR or PR to the first documentation of disease progression or death from any cause 36 months
Secondary Disease control rate (DCR) per RECIST version 1.1 according to investigator assessment (Phases 2 and 3) DCR is defined as subjects with CR, PR, or stable disease (SD or non-CR/non-progressive disease) 36 months
Secondary PFS per RECIST v1.1 according to blinded independent central review (BICR) assessment (Phase 3 only) PFS is defined as the time from randomization to the date of disease progression or death from any cause 36 months
Secondary Confirmed ORR per RECIST version 1.1 according to BICR assessment (Phase 3 only) ORR is defined as the proportion of subjects with best overall response of CR or PR 36 months
Secondary ORR per RECIST version 1.1 according to BICR assessment (Phase 3 only) ORR is defined as the proportion of subjects with best overall response of CR or PR 36 months
Secondary DOR per RECIST version 1.1 according to BICR assessment (Phase 3 only) DOR is defined as the time from first documentation of objective response of CR or PR to the first documentation of disease progression or death from any cause 36 months
Secondary DCR per RECIST version 1.1 according to BICR assessment (Phase 3 only) DCR is defined as subjects with CR, PR, or stable disease (SD or non-CR/non-progressive disease) 36 months
Secondary Incidence of AEs (Phase 3 only) An adverse event is any untoward medical occurrence in a subject or clinical investigational subject administered a medicinal product and which does not necessarily have a causal relationship with this treatment. 36 months
Secondary Incidence of laboratory abnormalities (Phase 3 only) To be summarized using descriptive statistics. 36 months
Secondary Incidence of dose modifications (Phase 3 only) 36 months
Secondary PFS per RECIST version 1.1 according to investigator assessment (Phase 2 only) PFS is defined as the time from the date of treatment initiation to the date of disease progression or death from any cause 18 months
Secondary Area under the plasma concentration-time curve (AUC) to the time of the last quantifiable concentration (AUClast) of tucatinib (Phase 2 only) Pharmacokinetic (PK) parameter 1 month
Secondary AUC to AUClast of paclitaxel (Phase 2 only) PK parameter 1 month
Secondary Maximum observed concentration (Cmax) of tucatinib (Phase 2 only) PK parameter 1 month
Secondary Cmax of paclitaxel (Phase 2 only) PK parameter 1 month
Secondary Time of Cmax (Tmax) of tucatinib (Phase 2 only) PK parameter 1 month
Secondary Tmax of paclitaxel (Phase 2 only) PK parameter 1 month
Secondary Trough concentration (Ctrough) of tucatinib (Phase 2 only) PK parameter 18 months
Secondary Ctrough of paclitaxel (Phase 2 only) PK parameter 18 months
Secondary Metabolic ratio of tucatinib based on AUC (MRAUC) (Phase 2 only) PK parameter 1 month
Secondary MRAUC of paclitaxel (Phase 2 only) PK parameter 1 month
Secondary Time to deterioration of GEC symptoms as assessed by the European Organisation for Research and Treatment or Cancer (EORTC) quality of life questionnaire (QLQ)-C30 and EORTC QLQ-OG25 questionnaires. The EORTC questionnaires measure aspects of health-related quality of life (HRQoL). Time to deterioration will be assessed in specific pre-specified single items from either the EORTC QLQ-C30 or EORTC QLQ OG25 and deterioration is defined as a 10-point increase from baseline in the symptom scales and a 10-point decrease from baseline for overall HRQoL. 36 months
Secondary Change from baseline in health-related quality of life (HRQoL) 36 months
Secondary Utility index values as assessed by the EQ-5D-5L The EQ-5D-5L questionnaire is used as a preference based measurement of HRQoL outcomes. Data will be summarized using descriptive statistics. 36 months
See also
  Status Clinical Trial Phase
Recruiting NCT05977998 - A Phase II Study of Perioperative Paclitaxel in Patients With Gastric Adenocarcinoma and Carcinomatosis or Positive Cytology Phase 2
Recruiting NCT05059444 - ORACLE: Observation of ResiduAl Cancer With Liquid Biopsy Evaluation
Not yet recruiting NCT04931420 - Study Comparing Standard of Care Chemotherapy With/ Without Sequential Cytoreductive Surgery for Patients With Metastatic Foregut Cancer and Undetectable Circulating Tumor-Deoxyribose Nucleic Acid Levels Phase 2
Recruiting NCT03257163 - Pembrolizumab, Capecitabine, and Radiation Therapy in Treating Patients With Mismatch-Repair Deficient and Epstein-Barr Virus Positive Gastric Cancer Phase 2
Completed NCT02128243 - Trial of S-1 Maintenance Therapy in Metastatic Esophagogastric Cancer Phase 2
Completed NCT01178944 - Pralatrexate and Oxaliplatin in Treating Patients With Unresectable or Metastatic Esophageal, Stomach, or Gastroesophageal Junction Cancer Phase 2
Terminated NCT00209079 - Phase II Trial of Gleevec and Taxotere as a Combined Regimen for Advanced Gastric Adenocarcinoma Phase 2
Terminated NCT02862535 - Study to Evaluate the Safety and Tolerability of Andecaliximab as Monotherapy and in Combination With Anti-Cancer Agents in Japanese Participants With Gastric or Gastroesophageal Junction Adenocarcinoma Phase 1
Active, not recruiting NCT05008783 - A Study of AK104 in the First-line Treatment of Locally Advanced Unresectable or Metastatic G/GEJ Adenocarcinoma Phase 3
Recruiting NCT04430738 - Tucatinib Plus Trastuzumab and Oxaliplatin-based Chemotherapy or Pembrolizumab-containing Combinations for HER2+ Gastrointestinal Cancers Phase 1/Phase 2
Recruiting NCT04114136 - Anti-PD-1 mAb Plus Metabolic Modulator in Solid Tumor Malignancies Phase 2
Completed NCT03196232 - Epacadostat and Pembrolizumab in Treating Patients With Metastatic or Unresectable Gastroesophageal Junction or Gastric Cancer Phase 2
Recruiting NCT04047953 - Paclitaxel (Albumin-bound) Combined With Oxaliplatin and S-1 Conversion Therapy for Gastric Adenocarcinoma N/A
Completed NCT02891447 - Heated Mitomycin and Cisplatin During Surgery in Treating Patients With Stomach or Gastroesophageal Cancer Phase 2
Completed NCT02864381 - Study to Evaluate the Efficacy and Safety of Andecaliximab Combined With Nivolumab Versus Nivolumab Alone in Adults With Unresectable or Recurrent Gastric or Gastroesophageal Junction Adenocarcinoma Phase 2
Terminated NCT04032704 - A Study of Ladiratuzumab Vedotin in Advanced Solid Tumors Phase 2
Terminated NCT04604132 - Derazantinib Alone or in Combination With Paclitaxel, Ramucirumab or Atezolizumab in Gastric Adenocarcinoma Phase 1/Phase 2
Completed NCT02830594 - Pembrolizumab and Palliative Radiation Therapy in Treating Patients With Metastatic Esophagus, Stomach, or Gastroesophageal Junction Cancer Phase 2
Recruiting NCT06038578 - A Study of TRK-950 When Used in Combination With Ramucirumab and Paclitaxel in Patients With Gastric Cancer Phase 2
Terminated NCT04099277 - A Study of LY3435151 in Participants With Solid Tumors Phase 1