Clinical Trials Logo

Clinical Trial Details — Status: Active, not recruiting

Administrative data

NCT number NCT02075840
Other study ID # BO28984
Secondary ID 2013-004133-33
Status Active, not recruiting
Phase Phase 3
First received
Last updated
Start date August 19, 2014
Est. completion date September 29, 2026

Study information

Verified date May 2024
Source Hoffmann-La Roche
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This randomized, active controlled, multicenter phase III open-label study is designed to evaluate the efficacy and safety of alectinib compared with crizotinib treatment in participants with treatment-naive anaplastic lymphoma kinase-positive (ALK-positive) advanced non-small cell lung cancer (NSCLC). Participants will be randomized in a 1:1 ratio to receive either alectinib, 600 milligrams (mg) orally twice daily (BID), or crizotinib, 250 mg orally BID. Participants will receive treatment until disease progression, unacceptable toxicity, withdrawal of consent, or death. The study is expected to last approximately 144 months.


Recruitment information / eligibility

Status Active, not recruiting
Enrollment 303
Est. completion date September 29, 2026
Est. primary completion date February 9, 2017
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: - Histologically or cytologically confirmed diagnosis of advanced or recurrent (Stage IIIB not amenable for multimodality treatment) or metastatic (Stage IV) NSCLC that is ALK-positive as assessed by the Ventana immunohistochemistry (IHC) test - Life expectancy of at least 12 weeks - Eastern cooperative oncology group performance status (ECOG PS) of 0-2 - Participants with no prior systemic treatment for advanced or recurrent (Stage IIIB not amenable for multimodality treatment) or metastatic (Stage IV) NSCLC - Adequate renal, and hematologic function - Participants must have recovered from effects of any major surgery or significant traumatic injury at least 28 days before the first dose of study treatment - Measurable disease by response evaluation criteria in solid tumors (RECIST) version 1.1 (v1.1) prior to the administration of study treatment - Prior brain or leptomeningeal metastases allowed if asymptomatic (e.g., diagnosed incidentally at study baseline) - Negative pregnancy test for all females of child bearing potential - Use of highly effective contraception as defined by the study protocol Exclusion Criteria: - Participants with a previous malignancy within the past 3 years - Any gastrointestinal (GI) disorder or liver disease - National cancer institute common terminology criteria for adverse events (NCI CTCAE) (version 4.0) Grade 3 or higher toxicities due to any prior therapy (e.g., radiotherapy) (excluding alopecia) - History of organ transplant - Co-administration of anti-cancer therapies other than those administered in this study - Participants with baseline QTc greater than (>) 470 milliseconds or symptomatic bradycardia - Recipient of strong/potent cytochrome P4503A inhibitors or inducers within 14 days prior to the first dose until the end of study treatment - Recipient of any drug with potential QT interval prolonging effects within 14 days prior to the first dose for all participants and while on treatment through the end of the study for crizotinib-treated participants only - History of hypersensitivity to any of the additives in the alectinib and crizotinib drug formulation - Pregnancy or lactation - Any clinically significant disease or condition (or history of) that could interfere with, or for which the treatment might interfere with, the conduct of the study or the absorption of oral medications or that would, in the opinion of the principal investigator, pose an unacceptable risk to the participant in this study - Any psychological, familial, sociological, or geographical condition potentially hampering compliance with the study protocol requirements and/or follow-up procedures; those conditions should be discussed with the participant before trial entry

Study Design


Intervention

Drug:
Alectinib
Participants will receive alectinib 600 mg orally (four 150 mg capsules) BID from Visit 0 (baseline) until disease progression, unacceptable toxicity, withdrawal of consent or death.
Crizotinib
Participants will receive crizotinib 250 mg capsules orally BID from Visit 0 (baseline) until disease progression, unacceptable toxicity, withdrawal of consent or death.

Locations

Country Name City State
Australia Kinghorn Cancer Centre; St Vincents Hospital Darlinghurst New South Wales
Australia Royal North Shore Hospital; Oncology St. Leonards New South Wales
Australia Monash Health Translational Precinct; Clinical Trials Centre, Level 3 Victoria
Australia Calvary Mater Newcastle; Medical Oncology Waratah New South Wales
Australia Queen Elizabeth Hospital; Medical Oncology Woodville South South Australia
Bosnia and Herzegovina University Clinical Centre of the Republic of Srpska; Clinic for Pulmonary Diseases Banja Luka
Bosnia and Herzegovina University Clinical Center Sarajevo;Clinic for Pulmonary disease Sarajevo
Bosnia and Herzegovina University Clinical Center Sarajevo;Institute of oncology Sarajevo
Brazil Hospital das Clinicas - UFRGS Porto Alegre RS
Brazil Instituto do Cancer do Estado de Sao Paulo - ICESP Sao Paulo SP
Canada Cross Cancer Institute Edmonton Alberta
Canada Saskatoon Cancer Centre; Uni of Saskatoon Campus Saskatoon Saskatchewan
Canada Mount Sinai Hospital; Oncology Toronto Ontario
Canada Sunnybrook Odette Cancer Centre Toronto Ontario
Chile Centro Internacional de Estudios Clínicos (CIEC) Recoleta
China Sun Yet-sen University Cancer Center Guangzhou City
China Shanghai Pulmonary Hospital Shanghai
Costa Rica Clinica CIMCA San José
Egypt Kasr Eieny Uni Hospital; Oncology (Nemrock) Cairo
France Chu Grenoble - Hopital Albert Michallon; Departement de Cancero-Hematologie Grenoble
France CHRU de Lille Lille
France Centre Leon Berard; Departement Oncologie Medicale Lyon
France Hopital Haut Leveque Pessac
France Hopital Pontchaillou Rennes
Germany St. Vincentius Kliniken Karlsruhe; Abteilung Hämatologie / Onkologie Karlsruhe
Germany Klinik Löwenstein gGmbH Medizinische Klinik II Löwenstein
Guatemala Grupo Angeles Guatemala City
Hong Kong Pamela Youde Nethersole Eastern Hospital; Clinical Oncology Hong Kong
Hong Kong Princess Margaret Hospital; Oncology Hong Kong
Hong Kong Queen Mary Hospital; Medicine & Respiratory Hong Kong
Hong Kong Tuen Mun Hospital; Clinical Oncology Hong Kong
Hong Kong Prince of Wales Hosp; Dept. Of Clinical Onc Shatin
Israel Rambam Medical Center; Oncology Haifa
Israel Meir Medical Center; Oncology Kfar-Saba
Italy Irccs Ist. Tumori Giovanni Paolo Ii; Dipartimento Oncologia Medica Bari Puglia
Italy Az Ospedaliera Nuovo Garibaldi Quartiere Nesima; Oncologia Medica Catania Sicilia
Italy Irccs Istituto Europeo Di Oncologia (IEO); Oncologia Medica Milano Lombardia
Italy Irccs Istituto Nazionale Dei Tumori (Int);S.C. Medicina Oncologica 1 Milano Lombardia
Italy Seconda Universita' Degli Studi; Divsione Di Oncologia Medica Napoli Campania
Italy Az. Osp. S. Luigi Gonzaga; Malattie Apparato Respiratorio 5 Ad Indirizzo Oncologico Orbassano Piemonte
Italy A.O. Universitaria Di Parma; Oncologia Medica Parma Emilia-Romagna
Italy Ospedale Provinciale Santa Maria Delle Croci; Oncologia Medica Ravenna Emilia-Romagna
Italy Policlinico Umberto i di Roma; dip. Scienze Radiologiche, Oncologiche, Anatomopatologiche Roma Lazio
Italy Azienda Ospedaliera Di Perugia Ospedale s. Maria Della Misericordia; Oncologia Medica Sant'Andrea Delle Fratte (PG) Umbria
Korea, Republic of National Cancer Center Goyang-si
Korea, Republic of Seoul National University Bundang Hospital Seongnam-si
Korea, Republic of Asan Medical Center 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
Mexico Instituto Nacional De Enfermedades Respiratorias;Unidad de Investigación Ciudad de México Mexico CITY (federal District)
New Zealand Uni of Auckland; Medical School Auckland
Poland Uniwersyteckie Centrum Kliniczne; Klinika Onkologii i Radioterapii Gdansk
Poland Ms Clinsearch Specjalistyczny Niepubliczny Zaklad Opieki Zdrowotnej Lublin
Poland Warminsko-Mazurskie Centrum Chorób P?uc w Olsztynie; Oddzial onkologii z pododdzialem chemioterapii Olsztyn
Poland Mazowieckie Centrum Leczenia Chorob Pluc I Gruzlicy; Oddzial Iii Otwock
Poland Centrum Onkologii - Inst.Im. Marii Sklodowskiej-Curie; Oncology Warszawa
Portugal CHUC - Unidade de Pneumologia Oncológica; Hospital de Dia de Oncologia Edificio Sao Jeronimo Coimbra
Portugal IPO de Lisboa; Servico de Pneumologia Lisboa
Portugal IPO do Porto; Servico de Oncologia Medica Porto
Russian Federation Moscow City Oncology Hospital #62 Moscovskaya Oblast Moskovskaja Oblast
Russian Federation N.N.Burdenko Main Military Clinical Hospital; Oncology Dept Moscow Moskovskaja Oblast
Russian Federation City Clinical Hospital No. 1 Novosibirsk
Russian Federation Medical Radiological Research Centre Rams; Dept. of Radiotherapy & Chemotherapy of Hemoblastosis Obninsk Kaluga
Russian Federation SPb City Clin Onc Dsp; Chemotherapy Sankt-peterburg Sankt Petersburg
Russian Federation Scientific Research Oncology Institute named after N.N. Petrov; Oncology St. Petersburg Sankt Petersburg
Serbia Clinical Center of Serbia Belgrade
Serbia Institute for pulmonary diseases of Vojvodina Sremska Kamenica
Singapore National Cancer Centre; Medical Oncology Singapore
Singapore National University Hospital; National University Cancer Institute, Singapore (NCIS) Singapore
Spain Hospital General Univ. de Alicante; Servicio de Oncologia Alicante
Spain Hospital Universitari Germans Trias i Pujol; Servicio de Oncologia Badalona Barcelona
Spain Hospital Universitario Quiron Dexeus Barcelona
Spain Hospital Universitario Puerta de Hierro; Servicio de Oncologia Majadahonda Madrid
Spain Hospital Univ Vall d'Hebron; Servicio de Oncologia Sant Andreu de La Barca Barcelona
Spain Hospital Universitario Virgen del Rocio; Servicio de Oncologia Sevilla
Switzerland Universitaetsspital Basel; Onkologie Basel
Switzerland Inselspital Bern; Universitätsklinik für Medizinische Onkologie, Klinische Forschungseinheit Bern
Switzerland CHUV; Departement d'Oncologie Lausanne
Switzerland UniversitätsSpital Zürich; Zentrum für Hämatologie und Onkologie, Klinik für Onkologie Zürich
Taiwan National Cheng Kung Univ Hosp Tainan
Taiwan National Taiwan University Hospital Taipei
Taiwan Taipei Veterans General Hospital Taipei City
Taiwan Taichung Veterans General Hospital Xitun Dist.
Thailand National Cancer Inst. Bangkok
Thailand Chiang Rai Prachanukroh Hospital; Department Of Medicine Chiang Rai
Thailand Khonkaen Hospital Khonkaen
Thailand King Chulalongkorn Memorial Hospital; Faculty of Medicine Chulalongkorn University Patumwan
Thailand Songklanagarind Hospital; Department of Internal Medicine, Division of Respiratory Songkhla
Turkey Baskent University Adana Dr. Turgut Noyan Practice and Research Hospital; Medical Oncology Adana
Turkey Ankara University Medical Faculty; Medikal Onkoloji Ankara
Turkey Trakya University Medical Faculty Research And Practice Hospital Medical Oncology Department Edirne
Turkey Inonu University Medical Faculty Turgut Ozal Medical Center Medical Oncology Department Malatya
Ukraine Dnipropetrovsk State Medical Academy; Chemotherapy Department Dnipropetrovsk
Ukraine Karkiv Regional Oncology Center Kharkiv
Ukraine Kyiv Regional Oncological Dispensary Kyiv
Ukraine Lviv State Oncology Regional Treatment and Diagnostic Centre; Department of hemotherapy Lviv
United Kingdom Birmingham Heartlands Hospital; Dept of Oncology Birmingham
United Kingdom Guys & St Thomas Hospital; Department of Oncology London
United Kingdom University College London Hospital London
United States Emory University Hospital Atlanta Georgia
United States University of Colorado Cancer Center Aurora Colorado
United States Beth Israel Deaconess Med Ctr; Hem/Onc Boston Massachusetts
United States Dana Farber Can Ins Boston Massachusetts
United States Massachusetts General Hospital Cancer Center Boston Massachusetts
United States University of Illinois at Chicago Chicago Illinois
United States UT Southwestern Medical Center Dallas Texas
United States University of Miami-Deerfield Beach Deerfield Beach Florida
United States Henry Ford Health System Detroit Michigan
United States Karmanos Cancer Institute Detroit Michigan
United States Banner MD Anderson Cancer Center Gilbert Arizona
United States St. Mary's Hospital Regional Cancer Center Grand Junction Colorado
United States University of Texas M.D. Anderson Cancer Center Houston Texas
United States Comprehensive Cancer Center - Peak Las Vegas Nevada
United States Northwest Georgia Oncology Centers, a Service of WellStar Cobb Hospital Marietta Georgia
United States SCRI Oncology Partners Nashville Tennessee
United States Columbia University Medical Center New York New York
United States North Valley Hem Onc Med Grp; Thomas&Dorothy Leavey Can Ctr Northridge California
United States Chao Family Comprehensive Cancer Center; UC Irvine Medical Center Orange California
United States Cancer Institute of Florida PA Orlando Florida
United States Memorial Health Care System Pembroke Pines Florida
United States Mayo Clinic Arizona Phoenix Arizona
United States TMPN/ Cancer Care Associates Redondo Beach California
United States Washington Uni School of Medicine Saint Louis Missouri
United States UCSF Helen Diller Family CCC San Francisco California

Sponsors (1)

Lead Sponsor Collaborator
Hoffmann-La Roche

Countries where clinical trial is conducted

United States,  Australia,  Bosnia and Herzegovina,  Brazil,  Canada,  Chile,  China,  Costa Rica,  Egypt,  France,  Germany,  Guatemala,  Hong Kong,  Israel,  Italy,  Korea, Republic of,  Mexico,  New Zealand,  Poland,  Portugal,  Russian Federation,  Serbia,  Singapore,  Spain,  Switzerland,  Taiwan,  Thailand,  Turkey,  Ukraine,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Progression-Free Survival (PFS) by Investigator Assessment PFS was assessed as time to disease progression or death whichever occurred first by investigator assessment using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (v1.1) Criteria. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 millimeter (mm) and the appearance of new lesions. Randomization to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Primary Percentage of Participants With PFS Event by Investigator Assessment PFS was assessed percentage of participants with disease progression or death whichever occurred first by investigator assessment using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (v1.1) Criteria. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 millimeter (mm) and the appearance of new lesions. Randomization to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Secondary PFS Independent Review Committee (IRC)-Assessed PFS was assessed as time to disease progression or death whichever occurred first by IRC assessment using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (v1.1) Criteria. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 mm and the appearance of new lesions. Randomization to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Secondary Percentage of Participants With PFS Event by IRC PFS was assessed as percentage of participants with disease progression or death whichever occurred first by IRC assessment using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (v1.1) Criteria. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 mm and the appearance of new lesions. Randomization to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Secondary Percentage of Participants With Central Nervous System (CNS) Progression as Determined by IRC Using RECIST V1.1 Criteria CNS progression was assessed as percentage of participants with an event defined as time from randomization until first radiographic evidence of CNS progression by IRC. The risk for a CNS progression without a prior non-CNS progression with alectinib compared with crizotinib. Randomization to CNS PD as first occurrence of disease progression (assessed every 8 weeks up to 33 months)
Secondary Percentage of Participants With Central Nervous System (CNS) Progression as Determined by IRC Using Revised Assessment in Neuro Oncology (RANO) Criteria CNS progression was assessed as percentage of participants with event defined as time from randomization until first radiographic evidence of CNS progression by IRC. The risk for a CNS progression without a prior non-CNS progression with alectinib compared with crizotinib. Randomization to the first occurrence of disease progression in the CNS (assessed every 8 weeks up to 33 months)
Secondary Percentage of Participants With Objective Response Rate (ORR) of Complete Response (CR) or Partial Response (PR) as Determined by The Investigators According to RECIST V1.1 Criteria ORR was defined as the percentage of participants who attained CR or PR. As per RECIST v1.1, CR: Disappearance of all target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm, PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters. Randomization to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Secondary Duration of Response (DOR) According to RECIST V1.1 Criteria as Assessed by the Investigators DOR was defined as the time from when response (CR or PR) was first documented to first documented disease progression or death, whichever occurred first. DOR was evaluated for participants who had a best overall response (BOR) of CR or PR. First occurrence of objective response to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Secondary Overall Survival (OS) Overall survival (OS) was defined as the time from randomization to death from any cause. From randomization until death (up to 43 months)
Secondary Percentage of Participants With OS Event Overall survival (OS) was defined as the time from randomization to death from any cause. From randomization until death (up to 43 months)
Secondary Percentage of Participants With CNS ORR of CR or PR IRC-assessed According to RECIST v1.1 Criteria CNS ORR was defined as the percentage of participants who attained CR or PR and had measurable/non-measurable CNS lesions at baseline. As per RECIST v1.1, CR: Disappearance of all target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm, PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters. Randomization to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Secondary CNS DOR IRC-assessed According to RECIST v1.1 Criteria CNS DOR was defined as the time from when response (CR or PR) was first documented to first documented disease progression or death, whichever occurred first. DOR was evaluated for participants who had a best overall response (BOR) of CR or PR. First occurrence of CNS objective response to first documented disease progression or death, whichever occurs first (assessed every 8 weeks up to 33 months)
Secondary Percentage of Participants With Adverse Events An adverse event (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. Baseline up to 28 months in the crizotinib arm and up to 30 months in the alectinib arm
Secondary Area Under The Concentration-Time Curve (AUC) of Alectinib Pre-dose (within 2 hours before alectinib) (baseline), 1, 2, 4, 6, and 8 hours post-dose at Visit 0 (first dosing day) and Week 4; Pre-dose (within 2 hours) at Week 8, then every 8 weeks until disease progression or death/withdrawal (up to 33 months)
Secondary Maximum Concentration (Cmax) of Alectinib Pre-dose (within 2 hours before alectinib), 1, 2, 4, 6, and 8 hours post-dose at baseline and Week 4; Pre-dose (within 2 hours before alectinib) at Week 8, then every 8 weeks until disease progression or death/withdrawal from study (up to 33 months)
Secondary Time to Reach Cmax (Tmax) of Alectinib Pre-dose (within 2 hours before alectinib), 1, 2, 4, 6, and 8 hours post-dose at baseline and Week 4; Pre-dose (within 2 hours before alectinib) at Week 8, then every 8 weeks until disease progression or death/withdrawal from study (up to 33 months)
Secondary AUC of Alectinib Metabolite Pre-dose (within 2 hours before alectinib) (baseline), 1, 2, 4, 6, and 8 hours post-dose at Visit 0 (first dosing day) and Week 4; Pre-dose (within 2 hours) at Week 8, then every 8 weeks until disease progression or death/withdrawal (up to 33 months)
Secondary Cmax of Alectinib Metabolite Pre-dose (within 2 hours before alectinib), 1, 2, 4, 6, and 8 hours post-dose at baseline and Week 4; Pre-dose (within 2 hours before alectinib) at Week 8, then every 8 weeks until disease progression or death/withdrawal from study (up to 33 months)
Secondary Tmax of Alectinib Metabolite Pre-dose (within 2 hours before alectinib), 1, 2, 4, 6, and 8 hours post-dose at baseline and Week 4; Pre-dose (within 2 hours before alectinib) at Week 8, then every 8 weeks until disease progression or death/withdrawal from study (up to 33 months)
Secondary Time to Deterioration by European Organization for The Research And Treatment of Cancer (EORTC) Quality Of Life Questionnaire Core 30 (C30) The EORTC QLQ-30 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Confirmed clinically meaningful deterioration in global health status or function is defined as a >or=10-point decrease from baseline in a symptom score that must be held for at least two consecutive assessments or an initial >or=10-point decrease from baseline followed by death within 5 weeks from the last assessment. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary Percentage of Participants With Deterioration by EORTC Quality Of Life Questionnaire Core 30 (C30) The EORTC QLQ-30 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Confirmed clinically meaningful deterioration in global health status or function is defined as a >or=10-point decrease from baseline in a symptom score that must be held for at least two consecutive assessments or an initial >or=10-point decrease from baseline followed by death within 5 weeks from the last assessment. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary Time to Deterioration by EORTC Quality of Life Questionnaire Lung Cancer Module 13 (LC13) The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Confirmed clinically meaningful deterioration in lung cancer symptoms is defined as a >or=10-point increase from baseline in a symptom score that must be held for at least two consecutive assessments or an initial >or=10-point increase above baseline followed by death within 5 weeks from the last assessment. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary Percentage of Participants With Deterioration by EORTC Quality of Life Questionnaire Lung Cancer Module 13 (LC13) The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Confirmed clinically meaningful deterioration in lung cancer symptoms is defined as a >or=10-point increase from baseline in a symptom score that must be held for at least two consecutive assessments or an initial >or=10-point increase above baseline followed by death within 5 weeks from the last assessment. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary Health-Related Quality of Life (HRQoL) by EORTC Quality of Life Questionnaire C30 Score The EORTC QLQ-C30 questionnaire consisted of 30 questions generating five functional scores (physical, role, cognitive, emotional, and social); a global health status/global quality of life scale score; three symptom scale scores (fatigue, pain, and nausea and vomiting); and six stand alone one-item scores that capture additional symptoms (dyspnea, appetite loss, sleep disturbance, constipation, and diarrhea) and perceived financial burden. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary HRQoL by EORTC Quality of Life Questionnaire LC13 Score Coughing The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary HRQoL by EORTC Quality of Life Questionnaire LC13 Score Dyspnoea The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary HRQoL by EORTC Quality of Life Questionnaire LC13 Score Pain in Chest The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Baseline, every 4 weeks until disease progression (up to 33 months)
Secondary HRQoL by EORTC Quality of Life Questionnaire LC13 Score Pain in Arm and Shoulder The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. All the scales and single-item scores were linearly transformed so that each score ranged from 0 to 100. A higher score on the global health and functioning subscales is indicative of better functioning. Baseline, every 4 weeks until disease progression (up to 33 months)
See also
  Status Clinical Trial Phase
Terminated NCT03087448 - Ceritinib + Trametinib in Patients With Advanced ALK-Positive Non-Small Cell Lung Cancer (NSCLC) Phase 1
Recruiting NCT05042375 - A Trial of Camrelizumab Combined With Famitinib Malate in Treatment Naïve Subjects With PD-L1-Positive Recurrent or Metastatic Non-Small Cell Lung Cancer Phase 3
Completed NCT02526017 - Study of Cabiralizumab in Combination With Nivolumab in Patients With Selected Advanced Cancers Phase 1
Enrolling by invitation NCT00068003 - Harvesting Cells for Experimental Cancer Treatments
Terminated NCT05414123 - A Therapy Treatment Response Trial in Patients With Leptomeningeal Metastases ((LM) Using CNSide
Recruiting NCT05059444 - ORACLE: Observation of ResiduAl Cancer With Liquid Biopsy Evaluation
Recruiting NCT05919537 - Study of an Anti-HER3 Antibody, HMBD-001, With or Without Chemotherapy in Patients With Solid Tumors Harboring an NRG1 Fusion or HER3 Mutation Phase 1
Recruiting NCT05009836 - Clinical Study on Savolitinib + Osimertinib in Treatment of EGFRm+/MET+ Locally Advanced or Metastatic NSCLC Phase 3
Recruiting NCT03412877 - Administration of Autologous T-Cells Genetically Engineered to Express T-Cell Receptors Reactive Against Neoantigens in People With Metastatic Cancer 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
Completed NCT03219970 - Efficacy and Safety of Osimertinib for HK Chinese With Metastatic T790M Mutated NSCLC-real World Setting.
Recruiting NCT05949619 - A Study of BL-M02D1 in Patients With Locally Advanced or Metastatic Non-small Cell Lung Cancer or Other Solid Tumors Phase 1/Phase 2
Recruiting NCT04054531 - Study of KN046 With Chemotherapy in First Line Advanced NSCLC Phase 2
Withdrawn NCT03519958 - Epidermal Growth Factor Receptor (EGFR) T790M Mutation Testing Practices in Hong Kong
Completed NCT03384511 - The Use of 18F-ALF-NOTA-PRGD2 PET/CT Scan to Predict the Efficacy and Adverse Events of Apatinib in Malignancies. Phase 4
Terminated NCT02580708 - Phase 1/2 Study of the Safety and Efficacy of Rociletinib in Combination With Trametinib in Patients With mEGFR-positive Advanced or Metastatic Non-small Cell Lung Cancer Phase 1/Phase 2
Completed NCT01871805 - A Study of Alectinib (CH5424802/RO5424802) in Participants With Anaplastic Lymphoma Kinase (ALK)-Rearranged Non-Small Cell Lung Cancer (NSCLC) Phase 1/Phase 2
Terminated NCT04042480 - A Study of SGN-CD228A in Advanced Solid Tumors Phase 1
Recruiting NCT05919641 - LIVELUNG - Impact of CGA in Patients Diagnosed With Localized NSCLC Treated With SBRT
Completed NCT03656705 - CCCR-NK92 Cells Immunotherapy for Non-small Cell Lung Carcinoma Phase 1