Clinical Trials Logo

Clinical Trial Details — Status: Terminated

Administrative data

NCT number NCT01641939
Other study ID # BO27952
Secondary ID 2012-000660-22
Status Terminated
Phase Phase 2/Phase 3
First received July 13, 2012
Last updated May 5, 2017
Start date September 3, 2012
Est. completion date April 30, 2016

Study information

Verified date May 2017
Source Hoffmann-La Roche
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This multicenter, randomized, adaptive Phase II/III study will evaluate the efficacy and safety of trastuzumab emtansine (T-DM1) compared to standard taxane (docetaxel or paclitaxel) treatment in participants with human epidermal growth factor receptor 2 (HER2)-positive advanced gastric cancer. At the start of the trial (stage 1), participants will be randomized with a ratio 2:2:1 to one of three treatment arms: Arm A: trastuzumab emtansine 3.6 milligram per kilogram (mg/kg) per intravenous injection (IV) every 3 weeks; Arm B: trastuzumab emtansine 2.4 mg/kg IV every week; Arm C: standard taxane therapy (docetaxel 75 milligram per meter square [mg/m^2] IV every 3 weeks or paclitaxel 80 mg/m^2 kg IV every week per investigator choice). At the end of the first stage of the study, the dose and schedule of trastuzumab emtansine that will be used in the second stage of the study will be selected by an Independent Data Monitoring Committee (IDMC). The regimen selection analysis will be made after approximately 100 participants across all three study arms have been treated for at least 12 weeks.

Once a trastuzumab emtansine regimen has been selected, Stage I participants who were assigned to the treatment arm which was selected for Stage II of the study and participants who were in the standard taxane group will continue to receive their assigned treatment regimen. Stage I participants who were assigned to the regimen that was not selected for further evaluation will continue to receive their assigned regimen and will continue to be followed for efficacy and safety. In Stage II of the study, additional participants will be recruited and randomized with a ratio 2:1 to either the selected regimen of trastuzumab emtansine or to the standard taxane therapy. Participants will receive study treatment until disease progression, unacceptable toxicity, initiation of another cancer therapy or withdrawal.


Recruitment information / eligibility

Status Terminated
Enrollment 415
Est. completion date April 30, 2016
Est. primary completion date June 30, 2015
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria:

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

- Life expectancy of at least 12 weeks from the first dose of study treatment

- Measurable and/or evaluable disease based on Response Evaluation Criteria in Solid Tumors (RECIST version 1.1)

- Adequate organ function as determined by the following laboratory results, within 28 days prior to randomization

- Participants must have a history of advanced gastric cancer (AGC), defined as unresectable and locally advanced or metastatic gastric cancer, including adenocarcinoma of the gastroesophageal junction (GEJ), and must have experienced disease progression during or after first-line therapy for their disease

- HER2-positive tumor (primary tumor or metastatic lesion) as confirmed by central laboratory HER2 testing (immunohistochemistry and/or in-situ hybridization)

- Participants must have received at least one prior chemotherapy regimen for AGC; prior therapy does not need to have included HER2-directed therapy.

- First-line therapy for AGC, including adenocarcinoma of the GEJ, must have included a combination of at least a platinum- and a fluoropyrimidine-based treatment given concurrently; prior therapy does not need to have included a HER2-directed therapy.

- Adjuvant or neoadjuvant therapy for AGC is allowed.

Exclusion Criteria:

- An interval shorter than 21 days from the last dose of chemotherapy or HER2-directed therapy until the time of randomization

- Prior treatment with trastuzumab emtansine, docetaxel, or paclitaxel either as single agents or as part of a treatment regimen.

- Treatment with any investigational anticancer drug within 21 days of the first study treatment administration

- More than one prior line of therapy for advanced gastric cancer

- History of other malignancy within the previous 5 years except for appropriately treated carcinoma in situ of the cervix, non-melanoma skin carcinoma, Stage I uterine cancer, or other malignancies with an expected curative outcome

- Brain metastases that are untreated or symptomatic or require any radiation, surgery, or steroid therapy to control symptoms from brain metastases within 1 month of randomization

- Peripheral neuropathy Grade >/=2

- Uncontrolled cardiopulmonary dysfunction (e.g., high blood pressure, serious cardiac arrhythmia)

- Other current, severe, uncontrolled systemic disease (e.g., clinically significant metabolic disease, wound healing disorders, ulcers)

- Clinically significant bleeding within 30 days before enrollment

- For female participants, current pregnancy or lactation

- Major surgical procedure or significant traumatic injury within 28 days prior to randomization or anticipation of the need for major surgery during the course of study treatment

- Infection with Human immunodeficiency virus (HIV) or hepatitis B virus, hepatitis C virus

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Taxane
Standard taxane (docetaxel 75 mg/m^2 IV every 3 weeks or paclitaxel 80 mg/m^2) IV once a week according to investigator choice.
trastuzumab emtansine
trastuzumab emtansine 3.6 mg/kg IV every 3 weeks
trastuzumab emtansine
trastuzumab emtansine 2.4 mg/kg IV once a week

Locations

Country Name City State
Argentina Fundación Investigar Buenos Aires
Argentina Hospital de Gastroenterologia Dr. Bonorino Udaondo ; Servicio de Oncología Buenos Aires
Argentina Instituto de Oncología de Rosario Rosario
Belgium UZ Leuven Gasthuisberg Leuven
Brazil Hospital de Cancer de Barretos Barretos SP
Brazil Hospital Amaral Carvalho Jau SP
Brazil Clinica de Oncologia de Porto Alegre - CliniOnco Porto Alegre RS
Brazil Hospital Sao Lucas - PUCRS Porto Alegre RS
Brazil Instituto Nacional de Cancer - INCa; Oncologia Rio de Janeiro RJ
Brazil Instituto do Cancer do Estado de Sao Paulo - ICESP Sao Paulo SP
Brazil Instituto de Oncologia de Sorocaba - CEPOS Sorocaba SP
Canada Brampton Memorial Hospital, William Osler Health Center Brampton Ontario
Canada St. Michael'S Hospital Toronto Ontario
Canada Toronto East General Hospital; Haematology/Oncology Toronto Ontario
Canada BCCA-Vancouver Cancer Centre Vancouver British Columbia
China Beijing Cancer Hospital Beijing
China The Affiliated Hospital of Military Medical Sciences(The 307th Hospital of Chinese PLA) Beijing
China Jilin Cancer Hospital Changchun
China the First Hospital of Jilin University Changchun
China Changzhou First People's Hospital Changzhou
China Third Affiliated Hospital of Third Military Medical University ChongQing
China Fujian Cancer Hospital Fuzhou
China Sun Yet-sen University Cancer Center Guangzhou
China Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou
China Harbin Medical University Cancer Hospital Harbin
China Jiangsu Cancer Hospital Nanjing
China The 81st Hospital of P.L.A. Nanjing
China Affiliated Hospital of Nantong University Nantong
China Fudan University Shanghai Cancer Center Shanghai
China Shanghai First People's Hospital Shanghai
China Zhongshan Hospital Fudan University Shanghai
China General Hospital of Shenyang Military Command of PLA Shenyang
China Union Hospital of Tongji Medical College, Dept. of Cancer Center; Cancer Center Wuhan
China First Affiliated Hospital of Medical College of Xi'an Jiaotong University Xi'an
China The Affiliated Hospital of Xuzhou Medical College Xuzhou
Czechia Fakultni Nemocnice Hradec Kralove; Dept of Radiotherapy & Oncology Hradec Kralove
Czechia Fakultni nemocnice Olomouc; Onkologicka klinika Olomouc
Czechia Fakultni Poliklinika Vseobecne Fakultni Niemocnice; Onkologicka Klinika Praha 2
Czechia Fakultní Nemocnice V Motole; Radioterapeuticko-Onkologicke Oddeleni Praha 5
Finland Tampere University Hospital; Dept of Oncology Tampere
France Hopital Augustin Morvan; Federation De Cancerologie Brest
France Hopital Beaujon; Gastro Enterologie 1 Clichy
France Centre Val Aurelle Paul Lamarque; Medecine A1 A2 Montpellier
France Hop Europeen Georges Pompidou; Gastro Enterologie Paris
France Hopital Saint Antoine; Hepatologie-Gastr-Enterologie Paris
France Hopital Robert Debre; Gastro Enterologie Reims
France Hopital Purpan; Unite Onco Digestive Toulouse
Germany Charité-Universitätsm. Berlin; Med. Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunolo. Berlin
Germany Universitätsklinikum "Carl Gustav Carus"; Med. Klinik & Poliklinik I, Arbeitsgr. intern. Onkologie Dresden
Germany Facharztzentrum Eppendorf, Studien GbR Hamburg
Germany Universitätsklinikum Köln Köln
Germany Tagesklinik Landshut; Hämatologie/Onkologie Landshut
Germany Onkologische Gemeinschaftspraxis Magdeburg
Guatemala Centro Oncológico Sixtino / Centro Oncológico SA Guatemala
Guatemala Grupo Angeles Guatemala City
Hungary Fovarosi Szent Laszlo Korhaz-Rendelointezet; Onkologiai Osztaly X Budapest
Hungary Szegedi Tudomanyegyetem, AOK, Szent-Gyorgyi Albert Klinikai Kozpont, Onkoterapias Klinika Szeged
Hungary Hetenyi Geza County Hospital; Onkologiai Kozpont Szolnok
Hungary Zala Megyei Kórház, Külsö Kórház, Pózva; Onkológiai Osztály Zalaegerszeg
Italy AZ.Osp S. Orsola - Malpighi-Reparto di Oncologia Medica Bologna Emilia-Romagna
Italy Campus Universitario S.Venuta; Centro Oncologico T.Campanella Catanzaro Calabria
Italy Azienda Ospedaliero-Universitaria Careggi;S.C. Oncologia Medica 1 Firenze Toscana
Italy A.O. Universitaria Pisana; Oncologia Pisa Toscana
Italy A.O. Città della Salute e della Scienza - Presidio Molinette; divisione oncologia medica Torino Piemonte
Japan Aichi Cancer Center Hospital; Clinical Oncology Aichi
Japan Chiba Cancer Center; Gastroenterology Chiba
Japan National Cancer Center Hospital East; Gastroenterology Chiba
Japan National Hospital Organization Shikoku Cancer Center; Gastroenterology Ehime
Japan Hokkaido University Hospital:Gastroenterology Hokkaido
Japan Hyogo Cancer Center; Gastroenterology Hyogo
Japan Hyogo College Of Medicine; Upper Gastroenterology Hyogo
Japan Ibaraki Prefectural Central Hospital; Gastroenterology Ibaraki
Japan Tohoku Uni Hospital; Clinical Oncology Miyagi
Japan Kindai University Hospital; Medical Oncology Osaka
Japan Osaka University Hospital; Surgery Osaka
Japan Saitama Cancer Center; Gastroenterology Saitama
Japan Shizuoka Cancer Center; Gastroenterology Shizuoka
Japan Shizuoka General Hospital; Clinical Oncology Shizuoka
Japan Tochigi Cancer Center; Medical Oncology Tochigi
Japan National Cancer Center Hospital; Gastrointestinal Oncology Tokyo
Japan The Cancer Institute Hospital, JFCR; Gastroenterology Tokyo
Japan Tokyo Metropolitan Komagome Hospital; Chemotherapy Tokyo
Japan Toranomon Hospital; Medical Oncology Tokyo
Korea, Republic of Asan Medical Center; Medical Oncology Seoul
Korea, Republic of Korea University Anam Hospital; Oncology Haemotology Seoul
Korea, Republic of Samsung Medical Center Seoul
Korea, Republic of Seoul National University Hospital Seoul
Korea, Republic of Seoul St.Mary's Hospital; Medical Oncology Seoul
Korea, Republic of Yonsei University Severance Hospital; Medical Oncology Seoul
Malaysia University Malaya Medical Centre; Clinical Oncology Unit, Kuala Lumpur
Malaysia Hospital Wanita dan Kanak-Kanak Sabah Sabah
Mexico Centenario Hospital Miguel Hidalgo Aguascalientes
Mexico Centro Estatal De Cancerologia De Chihuahua; Servicio De Hematologia Banco De Sangre Chihuahua
Mexico Hospital General de México; Unidad de Oncologia Mexico DF
Panama Centro Hemato Oncologico Paitilla Panama City
Peru Hospital Nacional Almanzor Aguinaga Asenjo; Unidad De Investigacion Del Servicio De Oncologia Medica Chiclayo
Peru Hospital Nacional Adolfo Guevara Velasco Cusco
Peru Hospital Nacional Edgardo Rebagliati Martins Jesus Maria
Peru Instituto Nacional de Enfermedades Neoplasicas Lima
Philippines Perpetual Succour Hospital Cebu
Philippines Veterans Memorial Medical Ctr; Cancer Research Centre Quezon City, Luzon
Poland Uniwersyteckie Centrum Kliniczne, Klinika Onkologii i Radioterapii Gdansk
Poland Szpital Uniwersytecki w Krakowie Krakow
Poland Wielkopolskie Centrum Onkologii; im. Marii Sklodowskiej-Curie Poznan
Poland Wojewódzki Szpital Specjalistyczny Nr 3 Rybnik
Poland Centrum Onkologii - Instytut im. M. Sklodowskiej-Curie; Klinika Gastroenterologii Onkologicznej Warszawa
Romania Institutul Clinic Fundeni Bucuresti Bucharest
Romania Medisprof SRL Cluj-Napoca
Romania Spitalul Universitar CF Cluj-Napoca; Sectia Oncologie Cluj-Napoca
Romania Spitalul Clinic Judetean Mures; Oncologie Medicala Targu Mures
Russian Federation Arkhangelsk Regional Clinical Oncology Dispensary Arkhangelsk
Russian Federation Ivanovo Regional Oncology Dispensary Ivanovo
Russian Federation Omsk Region Clinical Oncology Dispensary; 1St Sergical Department Omsk
Russian Federation State Budget Institution of Healthcare of Stavropol region Pyatigorsk Oncology Dispensary Pyatigorsk
Russian Federation Tula Regional Oncology Dispensary Tula
Singapore National Cancer Centre Singapore
Spain Hospital Clinic i Provincial; Servicio de Farmacia Barcelona
Spain Hospital General Universitario Gregorio Marañon; Servicio de Oncologia Madrid
Spain Hospital Universitario La Paz; Servicio de Oncologia Madrid
Spain Hospital Univ. Central de Asturias; Servicio de Oncologia Oviedo Asturias
Spain Hospital Universitario Marques de Valdecilla; Servicio de Oncologia Santander Cantabria
Spain Complejo Hospitalario Universitario de Santiago (CHUS) ; Servicio de Oncologia Santiago de Compostela La Coruña
Spain Hospital Universitario Virgen del Rocio; Servicio de Oncologia Sevilla
Spain Hospital Universitario Miguel Servet; Servicio Oncologia Zaragoza
Taiwan Kaohsiung Chang Gung Memorial Hospital; Dept of Hem and Onc Kaohsung
Taiwan National Taiwan Uni Hospital; Dept of Oncology Taipei
Taiwan Chang Gung Medical Foundation - Linkou; Division of Hematology- Oncology Taoyuan
Turkey Ataturk University Medical Faculty Yakutiye Research Hospital Medical Oncology Department Erzurum
Turkey Istanbul Bilim University School Of Medicine; Department Of Medical Oncology Istanbul
Turkey Marmara Uni Faculty of Medicine; Medical Oncology Istanbul
Turkey Ege Uni Medical Faculty; Oncology Dept Izmir
Turkey Hacettepe Uni Medical Faculty Hospital; Oncology Dept Sihhiye, ANKARA
United Kingdom Velindre Cancer Centre; Oncology Dept Cardiff
United Kingdom The Beatson West of Scotland Cancer Centre; Cancer Clinical Trials Unit Glasgow
United Kingdom Royal Marsden Hospital; Dept of Med-Onc London
United Kingdom Christie Hospital Nhs Trust; Medical Oncology Manchester
United Kingdom Royal Marsden Hospital; Dept of Medical Oncology Sutton
United Kingdom BRISTOL ONCOLOGY CENTRE; CLINICAL TRIALS UNIT; R & D department Weston Super Mare
United States Comprehensive Blood/Cancer Ctr Bakersfield California
United States Dana Farber Can Ins Boston Massachusetts
United States Massachusetts General Hospital. Boston Massachusetts
United States University of Texas M.D. Anderson Cancer Center Houston Texas
United States Norton Healthcare Inc. Louisville Kentucky
United States Vanderbilt Nashville Tennessee
United States Yale Cancer Center New Haven Connecticut
United States Weill Cornell Medical College New York New York
United States Stanford University School of Medicine Stanford California
United States University of Kansas; Medical Center & Medical pavilion Westwood Kansas

Sponsors (1)

Lead Sponsor Collaborator
Hoffmann-La Roche

Countries where clinical trial is conducted

United States,  Argentina,  Belgium,  Brazil,  Canada,  China,  Czechia,  Finland,  France,  Germany,  Guatemala,  Hungary,  Italy,  Japan,  Korea, Republic of,  Malaysia,  Mexico,  Panama,  Peru,  Philippines,  Poland,  Romania,  Russian Federation,  Singapore,  Spain,  Taiwan,  Turkey,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Overall Survival (OS)- Phase 3 Overall survival was defined as the time between the date of randomization and date of death due to any cause. Kaplan-Meier estimates were used for analysis. Participants for whom no death was reported prior to an analysis cutoff (30 June 2015) was censored at the latest date before the cutoff in which they were known to be alive. All data from the standard taxane therapy and trastuzumab emtansine 2.4 mg (selected treatment arm) from phase 2 and phase 3 (Stage 2) are combined into phase 3 data, and thus cumulative data are provided within the results presented for phase 3. The confirmatory analyses are restricted to comparisons between the taxane arm and the selected trastuzumab emtansine arm (2.4 mg). Date of randomization until death (up to 2 years 3 months)
Primary Overall Survival (OS) - Phase 2 (Dose Selection Portion of the Study) Overall survival was defined as the time between the date of randomization and date of death due to any cause. Kaplan-Meier estimates were used for analysis. Participants for whom no death was reported prior to an analysis cutoff (10 August 2013) was censored at the latest date before the cutoff in which they were known to be alive. Date of randomization until death (up to 1 year)
Secondary Percentage of Participants With Disease Progression or Death According to Modified Response Evaluation Criteria in Solid Tumors (mRECIST v1.1) - Phase 3 Progressive disease could base on symptom deterioration or was defined as at least a 20 percent (%) increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since treatment started or the appearance of one or more new lesions and/or the unequivocal progression of existing non-target lesions. Tumor assessment was performed using modified RECIST v1.1. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. Date of randomization until disease progression or death, whichever occurred first (assessed at baseline, every 6 weeks up to 2 years 3 months)
Secondary Progression Free Survival (PFS) According to Modified Response Evaluation Criteria in Solid Tumors (mRECIST v1.1) - Phase 3 Progression-free survival was defined as the time between the date of randomization and the first date of documented progression or date of death due to any cause, whichever occurred first. Tumor assessment was performed using modified RECIST v1.1. Progressive disease could base on symptom deterioration or was defined as at least a 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since treatment started or the appearance of one or more new lesions and/or the unequivocal progression of existing non-target lesions. Kaplan-Meier estimates were used for analysis. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. The confirmatory analyses are restricted to comparisons between the taxane arm and the selected trastuzumab emtansine arm (2.4 mg). Date of randomization until disease progression or death, whichever occurred first (assessed at baseline, every 6 weeks up to 2 years 3 months)
Secondary Percentage of Participants With Objective Response According to mRECIST v1.1 - Phase 3 Objective response referred to participants with complete response (CR) or partial response (PR). CR: disappearance of all target lesions, non-target lesions, and normalization of tumor marker level. PR: greater than or equal to (>=) 30% decrease in sum of the longest diameter (LD) of all target lesions taking as reference the screening sum LD. To be assigned a status of PR or CR, changes in tumor measurements had to be confirmed by repeat assessments that should have been performed no less than 4 weeks after the criteria for response were first met. Longer intervals as determined by the study protocol were also appropriate. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. Date of randomization until disease progression or death, whichever occurred first (assessed at baseline, every 6 weeks up to 2 years 3 months)
Secondary Duration of Objective Response (DOR) - Phase 3 DOR: time from the date when a clinical response [CR or PR] was first documented to the date of first documented progressive disease (PD) or death. CR:disappearance of all target lesions, non-target lesions, and normalization of tumor marker level. PR: >= 30% decrease in sum of the LD of all target lesions taking as reference the screening sum LD. PD: could base on symptom deterioration or at least a 20% increase in the sum of diameters of target or non-target lesions and new lesions, taking as reference the smallest sum on study (nadir), including baseline. To be assigned a status of PR or CR, changes in tumor measurements had to be confirmed by repeat assessments that should have been performed no less than 4 weeks after the criteria for response were first met. Longer intervals as determined by the study protocol were also appropriate. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. Date of randomization until disease progression or death, whichever occurred first (assessed at baseline, every 6 weeks up to 2 years 3 months)
Secondary Percentage of Participants With Clinically Significant Improvement in European Organisation for Research and Treatment of Cancer Quality of Life Core Module 30 (EORTC QLQ-C30) Score - Phase 3 The EORTC QLQ-C30 is a validated, cancer-specific 30-item patient-reported measure, and contains 14 domains to assess the impact of cancer treatment on 5 aspects of participants functioning (physical, role, cognitive, emotional, and social), 9 aspects of disease/treatment-related symptoms (fatigue, nausea and vomiting, pain, dyspnea, insomnia, loss of appetite, constipation, diarrhea) and a global QoL/overall health status scale. Questions used 4 point scale (1 'Not at all' to 4 'Very much'; with the exception of the QoL/health status scale which uses a 7-point scale (1 'very poor' to 7 'Excellent'). Each scale is transformed on a scale of 0-100; a higher score equals (=) a better level of functioning or greater degree of symptoms. Change of greater than or equal to (>=) 10-points has been found to be clinically significant. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. Day 1 of each treatment cycle, at the study drug completion visit, and thereafter at follow-up (up to 2 years 3 months)
Secondary Percentage of Participants With Clinically Significant Improvement in Quality of Life Questionnaire Stomach Cancer Module 22 (QLQ-STO22) Score - Phase 3 The Quality of Life Questionnaire Stomach Cancer Module 22 (QLQ-STO22) supplements the EORTC QLQ-C30 to assess symptoms and treatment-related side effects commonly reported in participants. There are 22 questions which comprise 5 scales (dysphagia, pain, reflux symptom, dietary restrictions, and anxiety) and 4 single items (dry mouth, hair loss, taste, body image). Most questions use 4-point scale (1 'Not at all' to 4 'Very much'; 1 question was a yes or no answer). A linear transformation was used to standardize all scores and single-items to a scale of 0 to 100; higher score=better level of functioning or greater degree of symptoms. Change of >=10 points has been found to be clinically significant. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. Day 1 of each treatment cycle, at the study drug completion visit, and thereafter at survival follow-up (up to 2 years 3 months)
Secondary Percentage of Participants With Advanced Gastric Cancer (AGC) Symptom Progression - Phase 3 AGC symptomatic progression: a worsening of >=10-points in any 1 of the abdominal discomfort, loss of appetite, weakness and fatigue, upper abdominal pain, change in bowel movement, and/or weight loss scales of the EORTC QLQ-C30 and QLQ-STO22. QLQ-STO22 supplements EORTC QLQ-C30 to assess symptoms and commonly reported treatment-related side effects. There are 22 questions comprise 5 scales (dysphagia, pain, reflux symptom, diet restrictions, anxiety), 4 single items (dry mouth, hair loss, taste, body image), which are related to the symptoms of the disease. Most questions used 4-point scale (1 'Not at all' to 4 'Very much'). All scores and single-items transformed to a scale of 0-100; higher score=better level of functioning or greater degree of symptoms. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. Day 1 of each treatment cycle, at the study drug completion visit, and thereafter at survival follow-up (up to 2 years 3 months)
Secondary Time to Advanced Gastric Cancer (AGC) Symptom Progression - Phase 3 Time to AGC symptom were defined as the time from randomization to the first documentation of an increase in at least one of the pre-specified abdominal discomfort, loss of appetite, weakness and fatigue, upper abdominal pain, change in bowel movement, and weight loss subscales of the QLQ STO22 and EORTC QLQ-C30. Cumulative data (up to primary analysis cut-off date of 30-June-2015) are provided for both phase 2 and phase 3 within the results of this measure. Day 1 of each treatment cycle, at the study drug completion visit, and thereafter at survival follow-up (up to 2 years 3 months)
Secondary Maximum Observed Plasma Concentration (Cmax) of Trastuzumab Emtansine (T-DM1) and Total Trastuzumab - Stage 1 Maximum observed plasma concentration of Trastuzumab Emtansine (T-DM1) and total trastuzumab were reported. Stage 1 consists of all participants recruited before the regimen selection, which was carried out after 12 weeks of randomization. Day 1 (D1) of Cycle 1 (C1) and C4, C1D2, C1D3, C1D4/C1D5, C1D8, C1D15, C2D1 (up to 12 weeks)
Secondary Maximum Observed Plasma Concentration (Cmax) of N2'-Deacetyl-N2'-(3-mercapto-1-oxopropyl)-Maytansine (DM1) - Stage 1 Maximum observed plasma concentration of DM1 were reported. Stage 1 consists of all participants recruited before the regimen selection decision. Regimen selection analysis was carried out after 12 weeks of randomization. C1D1 and C1C4, C1D2, C1D3, C1D4/C1D5, C1D8, C1D15, C2D1 (up to 12 weeks)
Secondary Maximum Observed Plasma Concentration (Cmax) of Trastuzumab Emtansine (T-DM1) and Total Trastuzumab - Stage 2 Stage 2 consists of all participants recruited after the regimen selection decision up to primary data cut-off date 30-June-2015. C1D1; C4D1
Secondary Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUCinf] - Stage 1 AUCinf= Area under the plasma concentration versus time curve (AUC) from time zero (pre-dose) to extrapolated infinite time (0 - inf). It is obtained from AUC (0 - t) plus AUC (t - inf). Stage 1 consists of all participants recruited before the regimen selection decision. Regimen selection analysis was carried out after 12 weeks of randomization. D1C1 and D1C4, C1D2, C1D3, C1D4/C1D5, C1D8, C1D15, C2D1 (up to 12 weeks)
Secondary Plasma Decay Half-Life (t1/2) - Stage 1 Plasma decay half-life is the time measured for the plasma concentration to decrease by one half. Stage 1 consists of all participants recruited before the regimen selection decision. Regimen selection analysis was carried out after 12 weeks of randomization. D1C1 and D1C4, C1D2, C1D3, C1D4/C1D5, C1D8, C1D15, C2D1 (up to 12 weeks)
Secondary Volume of Distribution at Steady State (Vss) - Stage 1 Volume of distribution is defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired blood concentration of a drug. Steady state volume of distribution (Vss) is the apparent volume of distribution at steady-state. Stage 1 consists of all participants recruited before the regimen selection decision. Regimen selection analysis was carried out after 12 weeks of randomization. D1C1 and D1C4, C1D2, C1D3, C1D4/C1D5, C1D8, C1D15, C2D1 (up to 12 weeks)
Secondary Systemic Clearance (CL) - Stage 1 CL is a quantitative measure of the rate at which a drug substance is removed from the body. Stage 1 consists of all participants recruited before the regimen selection decision. Regimen selection analysis was carried out after 12 weeks of randomization. D1C1 and D1C4, C1D2, C1D3, C1D4/C1D5, C1D8, C1D15, C2D1 (up to 12 weeks)
See also
  Status Clinical Trial Phase
Recruiting NCT05551416 - The EpiGASTRIC/EDGAR Project: New Strategies for the Early Detection and Prevention of Gastric Cancer
Recruiting NCT05518929 - Hypoxia During Gastroenterological Endoscope Procedures Sedated With Ciprofol In Overweight Or Obesity Patients Phase 4
Recruiting NCT06006390 - CEA Targeting Chimeric Antigen Receptor T Lymphocytes (CAR-T) in the Treatment of CEA Positive Advanced Solid Tumors Phase 1/Phase 2
Recruiting NCT03219593 - Apatinib as the First-Line Therapy in Elderly Locally Advanced or Metastatic Gastric Cancer Phase 2
Recruiting NCT05489211 - Study of Dato-Dxd as Monotherapy and in Combination With Anti-cancer Agents in Patients With Advanced Solid Tumours (TROPION-PanTumor03) Phase 2
Recruiting NCT05536102 - The Effectiveness and Safety of XELOX and Tislelizumab + PLD for Resectable Gastric Cancer (LidingStudy) Phase 2
Active, not recruiting NCT03170960 - Study of Cabozantinib in Combination With Atezolizumab to Subjects With Locally Advanced or Metastatic Solid Tumors Phase 1/Phase 2
Recruiting NCT06010862 - Clinical Study of CEA-targeted CAR-T Therapy for CEA-positive Advanced/Metastatic Malignant Solid Tumors Phase 1
Recruiting NCT05415098 - Study of Safety, Pharmacokinetic and Efficacy of APG-5918 in Advanced Solid Tumors or Lymphomas Phase 1
Active, not recruiting NCT04082364 - Combination Margetuximab, Retifanlimab, Tebotelimab, and Chemotherapy Phase 2/3 Trial in HER2+ Gastric/GEJ Cancer Phase 2/Phase 3
Withdrawn NCT03766607 - Trastuzumab Beyond Progression in HER2 Positive Metastatic Gastric Cancer Phase 2
Recruiting NCT04118114 - Phase II Study of PRL3-ZUMAB in Advanced Solid Tumors Phase 2
Completed NCT01924533 - Efficacy and Safety Study of Olaparib in Combination With Paclitaxel to Treat Advanced Gastric Cancer. Phase 3
Recruiting NCT05107674 - A Study of NX-1607 in Adults With Advanced Malignancies Phase 1
Active, not recruiting NCT04908813 - Study of HLX22 in Combanition With Trastuzumab and Chemotherapy Versus Placebo in Combination With Trastuzumab and Chemotherapy for Treatment of Locally Advanced or Metastatic Gastric Cancer Phase 2
Active, not recruiting NCT04249739 - Pembrolizumab + Capecitabine/Oxaliplatin (CapeOx) -HER2 Nagative and Pembrolizumab + Trastuzumab + Cisplatin/Capecitabine HER2 Positive Phase 2
Recruiting NCT05514158 - To Evaluate the Safety, Tolerability, Pharmacokinetics and Preliminary Efficacy of Disitamab Vedotin Combined With RC98 in the Treatment of Subjects With HER2-expressing Locally Advanced or Metastatic Gastric Cancer (Including AEG) Phase 1
Recruiting NCT04931654 - A Study to Assess the Safety and Efficacy of AZD7789 in Participants With Advanced or Metastatic Solid Cancer Phase 1/Phase 2
Recruiting NCT03175224 - APL-101 Study of Subjects With NSCLC With c-Met EXON 14 Skip Mutations and c-Met Dysregulation Advanced Solid Tumors Phase 2
Recruiting NCT05483491 - KK-LC-1 TCR-T Cell Therapy for Gastric, Breast, Cervical, and Lung Cancer Phase 1