Phase II Safety and Efficacy Study of Crizotinib in East Asian Patients With ROS1 Positive, ALK Negative Advanced NSCLC

Sponsor
Pfizer (Industry)
Overall Status
Completed
CT.gov ID
NCT01945021
Collaborator
OxOnc Development LP (Other)
129
44
1
75.7
2.9
0

Study Details

Study Description

Brief Summary

To assess treatment effectiveness and safety of oral crizotinib administered to East Asian patients with Advanced Non-Small Cell Lung Cancer (NSCLC) that is confirmed to be positive for a ROS1 positive gene mutation (translocation or inversion) and confirmed negative for an ALK mutation

Condition or Disease Intervention/Treatment Phase
Phase 2

Study Design

Study Type:
Interventional
Actual Enrollment :
129 participants
Allocation:
N/A
Intervention Model:
Single Group Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
Phase II, Open Label, Single Arm Study of the Efficacy and Safety of Crizotinib in East Asian Patients With Advanced ALK-Negative NSCLC Harboring a Translocation or Inversion Involving the c-ROS Oncogene (ROS1) Locus
Actual Study Start Date :
Sep 30, 2013
Actual Primary Completion Date :
Jul 31, 2015
Actual Study Completion Date :
Jan 22, 2020

Arms and Interventions

Arm Intervention/Treatment
Experimental: Crizotinib

Single-arm trial whereby all consented, enrolled, eligible patients receive crizotinib

Drug: Crizotinib
Other Names:
  • Xalkori
  • Outcome Measures

    Primary Outcome Measures

    1. Independent Radiology Reviewed Overall Objective Response (ORR) [Starting from the first dose study treatment until the first documented CR or PR (every 8 weeks then after 8 cycles at every 12 weeks in duration of 94.0 weeks)]

      Overall objective response (ORR) was defined as the number of patients with a best overall response of confirmed Complete Response or confirmed Partial Response according to RECIST v1.1 (as determined by Independent Radiology Review [IRR]), relative to the total population of response-evaluable participants. Per RECIST v1.1, CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); PR: at least 30% decrease in sum of diameters of target lesions, taking as reference baseline sum diameters. Confirmed responses were those that persisted on repeat imaging at least 4 weeks after the initial documentation of response.

    Secondary Outcome Measures

    1. IRR-Assessed Duration of Response (DR) [From first documentation of objective tumor response to first documentation of objective PD or death due to any cause, whichever occurred first (every 8 weeks then after 8 cycles at every 12 weeks in duration of 151.3 weeks)]

      DR: time from first documentation of objective tumor response (CR or PR) to first documentation of objective progressive disease (PD) or to death due to any cause, whichever occurred first. If no progression or death on study was observed, or given antitumor treatment other than study drug, participants were censored on date of last on-study tumor assessment. RECIST v1.1, a) PD: >=20% increase in sum of diameters of target lesions, taking as reference the smallest sum on study (this includes baseline sum if that is the smallest on study), sum must also demonstrate an absolute increase of >=5 mm, appearance of 1 or more new lesions, unequivocal progression of existing non-target lesions; b) CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); c) PR: >=30% decrease in sum of diameters of target lesions, taking as reference baseline sum diameters.

    2. IRR-Assessed Time to Tumor Response (TTR) [From date of first dose of crizotinib to first documentation of objective response was observed (every 8 weeks then after 8 cycles at every 12 weeks in duration of 151.3 weeks)]

      TTR was defined as the time from the date of first dose to first documentation of objective tumor response (CR or PR), that was subsequently confirmed. For participants proceeding from PR to CR, the onset of PR was taken as the onset of response. RECIST v1.1 (as determined by IRR), a) CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); b) PR: at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.

    3. IRR Assessed Disease Control Rate (DCR) at 8 Weeks [At 8 weeks after the start of study treatment]

      DCR at 8 weeks was defined as the percentage of participants with a confirmed CR, confirmed PR, or stable disease (SD) at 8 weeks, respectively, relative to the total population of response evaluable participants. RECIST v1.1 (as determined by IRR), a) CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); b) PR: at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study.

    4. IRR-Assessed Progression Free Survival (PFS) [From the date of first dose of crizotinib until the first documentation of objective PD or death (every 8 weeks then after 8 cycles at every 12 weeks in duration of 151.3 weeks)]

      PFS was defined as the time from the date of the first dose of crizotinib to first documentation of objective PD or to death on study due to any cause, whichever occurred first. If no progression or death on study was observed, or given antitumor treatment other than study drug, participants were censored on date of last on-study tumor assessment. RECIST v1.1, PD: at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered a sign of progression. Unequivocal progression of existing non-target lesions.

    5. Overall Survival (OS) [From date of the first dose of crizotinib until the date of death from any cause (up to 291.9 weeks)]

      OS was defined as the time from the date of the first dose of crizotinib to the date of death due to any cause. For participants still alive at the time of analysis, the OS time was censored on the last date the participants were known to be alive.

    6. Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious AEs, Treatment Emergent Treatment Related AEs and SAEs, Grade 3 or 4 Treatment Emergent AEs and Grade 3 or 4 Treatment Emergent Treatment Related AEs [Baseline up to 28 days after the last dose of study treatment (maximum up to 295.9 weeks)]

      Treatment-emergent AEs :between first dose of study drug and up to 28 days after last dose that were absent before treatment or that worsened relative to pretreatment state. Relatedness to crizotinib was assessed by the investigator. Treatment-related AE: any untoward medical occurrence attributed to study drug in a participant who received study drug. Serious adverse event (SAE):an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. Grade 3 (Severe) events=unacceptable or intolerable events, significantly interrupting usual daily activity, require systemic drug therapy/other treatment. Grade 4 (Life-threatening) events caused participant to be in imminent danger of death.

    7. Number of Participants With a Shift in Hematology Laboratory Results From Baseline Grade </=2 to Worst Grade 3 or Grade 4 [Baseline up to 28 days after the last dose of study treatment (maximum up to 295.9 weeks)]

      Laboratory values included hemoglobin increased, anemia, platelet count decreased, leukocytosis, white blood cell decreased, lymphocyte count increased, lymphocyte count decreased and neutrophil count decreased. Laboratory values were defined as National Cancer Institute Common Toxicity Criteria for Adverse Events Version 3.0 (NCI CTCAE v3.0) grade 3 or higher.

    8. Number of Participants With a Shift of Chemistry Laboratory Results From Baseline Grade </=2 to Worst Grade 3 or Grade 4 [Baseline up to 28 days after the last dose of study treatment (maximum up to 295.9 weeks)]

      Laboratory values included blood bilirubin increased, alanine aminotransferase increased, aspartate aminotransferase increased, alkaline phosphatase increased, hypoalbuminemia, hypernatremia, hyponatremia, hyperkalemia, hypokalemia, hypercalcemia, hypocalcemia, hypophosphatemia, Creatinine increased, hyperuricemia, hypermagnesemia, hypomagnesemia, hyperglycemia and hypoglycemia. Laboratory values were defined as National Cancer Institute Common Toxicity Criteria for Adverse Events Version 3.0 (NCI CTCAE v3.0) grade 3 or higher.

    9. Change From Baseline to Cycle 60 in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life (QOL) Questionnaire Core 30 (QLQ-C30) Scores [Baseline up to Cycle 60]

      The EORTC QLQ-C30 consists of 30 questions which are incorporated into 5 functional domains (physical, role, cognitive, emotional, and social); a global health status/global QOL scale; 3 symptom scales (fatigue, pain, nausea and vomiting scales); and 6 single items that assess the additional symptoms (dyspnea, appetite loss, sleep disturbance/insomnia, constipation, and diarrhea) and the perceived financial burden of treatment. All the scales and single-item scores ranged from 0 to 100, higher score is indicative of a higher response level (high score for a functional scale represents a high / healthy level of functioning; high score for the global health status / QoL represents a high QoL; a high score for a symptom scale / item represents a high level of symptomatology / problems).

    10. Change From Baseline to Cycle 60 in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Lung Cancer Module 13 Scores [Baseline up to Cycle 60]

      The EORTC QLQ-LC13 consisted of 1 multi-item scale and 9 single items that assessed specific symptoms (dyspnea, cough, hemoptysis, and site-specific pain), side effects (sore mouth, dysphagia, neuropathy, and alopecia). Scores on each scale and item ranged from 0 to 100, higher score is indicative of a higher response level (a high score for a symptom scale / item represents a high level of symptomatology / problems).

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    18 Years and Older
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    • Histologically or cytologically proven diagnosis of NSCLC that is locally advanced or metastatic

    • treatment-naïve or have received no more than 3 systemic treatment regimen(s)

    • Positive for translocation or inversion events involving the ROS1 gene

    • Negative for translocation or inversion events involving the ALK gene

    • Patients with brain metastases are eligible if asymptomatic, or if treated, must be neurologically stable for at least 2 weeks and are not taking any contraindicated medications

    • Any prior treatment (chemotherapy, radiation [except for palliative], or surgery) must have been completed at least 2 weeks prior to initiation of study medication

    • At least 1 measurable tumor lesion as per RECIST v1.1

    • Female or male, 18 years of age or older

    • ECOG performance status 0 to 1

    • Adequate organ function

    • Signed and dated informed consent

    • Willingness and ability to comply with scheduled visits, treatment plans, laboratory tests, and other study procedures, including completion of the PRO measures

    • Agree to use effective contraception during the study period and for at least 90 days after completion of the study treatment

    Exclusion Criteria:
    • Current treatment on another therapeutic clinical trial

    • Prior therapy specifically directed against ALK or ROS1 fusion genes

    • Spinal cord compression unless treated with the patient attaining good pain control and stable or recovered neurologic function, carcinomatous meningitis, or leptomeningeal disease

    • known interstitial fibrosis or interstitial lung disease

    • myocardial infarction, severe/unstable angina, coronary/peripheral artery bypass graft, congestive heart failure, or cerebrovascular accident including transient ischemic attack within 3 months prior to start of study treatment

    • Ongoing cardiac dysrhythmias of NCI CTCAE v4.03 Grade >/=2, uncontrolled atrial fibrillation of any grade, or QTc >470 msec

    • Pregnant or breast feeding

    • Use of drugs or foods that are known potent CYP3A4 inhibitors or inducers

    • Use of other anti-cancer drugs including traditional Chinese medicine on the SFDA list

    • Evidence of active malignancy within last 3 years

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 The First Affiliated Hospital of Anhui Medical University, Department of Medical Oncology Hefei Anhui China 230022
    2 Department of Medical Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences Chaoyang District Beijing China 100021
    3 The Military General Hospital of Beijing PLA / Medical Oncology Dept. Dongcheng District Beijing China 100700
    4 307 Hospital of PLA/Department of Lung Cancer Fengtai District Beijing China 100071
    5 Beijing Cancer Hospital, Department of Thoracic Oncology Haidian District Beijing China 100142
    6 Chinese PLA General Hospital Haidian District Beijing China 100853
    7 Beijing Chest Hospital Tongzhou District Beijing China
    8 Respiration department,the First Affiliated Hospital of Third Military Medical University, PLA Shapingba District Chongqing China 400038
    9 Fujian Province Oncology Hospital Fuzhou Fujian China 350014
    10 SUN Yat-Sen University Cancer Center Guangzhou Guangdong China 510060
    11 The First Affiliated Hospital of Guangzhou Medical College Guangzhou Guangdong China 510120
    12 Guangdong General Hospital Guangzhou Guangdong China
    13 Hunan Provincial Tumor Hospital/Division of Oncology Changsha Hunan China 410013
    14 Department of Oncology, Jilin Provincial Cancer Hospital Changchun Jilin China
    15 The affiliated hospital of medical college Qingdao University / Medical oncology department Qingdao Shandong China 266101
    16 Department of Pulmonary Medicine, Shanghai Chest Hospital Shanghai Shanghai China 200030
    17 Shanghai Chest Hospital/Lung cancer clinical center Shanghai Shanghai China 200030
    18 Shanghai Pulmonary Hospital Shanghai Shanghai China 200433
    19 Zhongshan Hospital Fudan University / Respiratory Department Xuhui District Shanghai China 200032
    20 Oncology Department, West China Hospital of Sichuan University Chengdu Sichuan China 610041
    21 Sichuan Province Cancer Hospital/Department of Pulmonary Tumor Chengdu Sichuan China 610041
    22 Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital Hexi District Tianjin China 300060
    23 The First Affiliated Hospital of College of Medicine, Zhejiang University Hangzhou Zhejiang China 310003
    24 Sir Run Run Shaw Hospital of College of Medicine of Zhejiang University, Center for Oncology Hangzhou Zhejiang China 310016
    25 Zhejiang Cancer Hospital Hangzhou Zhejiang China
    26 Shizuoka Cancer Center Sunto-gun Shizuoka Japan 411-8777
    27 Aichi Cancer Center Hospital Aichi Japan
    28 National Cancer Center Hospital East Chiba Japan
    29 NHO Shikoku Cancer Center Ehime Japan
    30 NHO Kyushu Cancer Center Fukuoka Japan
    31 Hyogo Cancer Center Hyogo Japan
    32 Tohoku University Hospital Miyagi Japan
    33 Kinki University Hospital Osaka Japan
    34 Osaka City General Hospital Osaka Japan
    35 Cancer Institute Hospital of JFCR Tokyo Japan
    36 National Cancer Center Hospital Tokyo Japan
    37 Asan Medical Center Seoul Korea, Republic of
    38 Samsung Medical Center Seoul Korea, Republic of
    39 Seoul National University Hospital Seoul Korea, Republic of
    40 Yonsei University, Severance Hospital Seoul Korea, Republic of
    41 Taichung Veterans General Hospital Taichung Taiwan
    42 National Cheng Kung University Hospital Tainan Taiwan
    43 National Taiwan University Hospital Taipei Taiwan
    44 Taipei Veterans General Hospital Taipei Taiwan

    Sponsors and Collaborators

    • Pfizer
    • OxOnc Development LP

    Investigators

    • Study Director: Pfizer CT.gov Call Center, Pfizer

    Study Documents (Full-Text)

    None provided.

    More Information

    Publications

    None provided.
    Responsible Party:
    Pfizer
    ClinicalTrials.gov Identifier:
    NCT01945021
    Other Study ID Numbers:
    • OO 12-01
    First Posted:
    Sep 18, 2013
    Last Update Posted:
    Feb 21, 2021
    Last Verified:
    Feb 1, 2021

    Study Results

    Participant Flow

    Recruitment Details
    Pre-assignment Detail 129 participants with anaplastic lymphoma kinase (ALK) negative advanced non-small cell lung cancer (NSCLC) harboring a translocation or inversion event involving the c-ros oncogene 1 (ROS1) locus were enrolled of whom 127 were allocated to treatment with crizotinib and 2 participants had screen failures.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Period Title: Overall Study
    STARTED 127
    Safety Population 127
    Response Evaluable Population 127
    Patient Reported Outcome Evaluable 123
    COMPLETED 44
    NOT COMPLETED 83

    Baseline Characteristics

    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Overall Participants 127
    Age (Years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [Years]
    52.48
    (12.136)
    Sex: Female, Male (Count of Participants)
    Female
    73
    57.5%
    Male
    54
    42.5%
    Race/Ethnicity, Customized (Count of Participants)
    Asian
    127
    100%
    Region of Enrollment (Count of Participants)
    China
    74
    58.3%
    Japan
    26
    20.5%
    Korea
    12
    9.4%
    Taiwan
    15
    11.8%

    Outcome Measures

    1. Primary Outcome
    Title Independent Radiology Reviewed Overall Objective Response (ORR)
    Description Overall objective response (ORR) was defined as the number of patients with a best overall response of confirmed Complete Response or confirmed Partial Response according to RECIST v1.1 (as determined by Independent Radiology Review [IRR]), relative to the total population of response-evaluable participants. Per RECIST v1.1, CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); PR: at least 30% decrease in sum of diameters of target lesions, taking as reference baseline sum diameters. Confirmed responses were those that persisted on repeat imaging at least 4 weeks after the initial documentation of response.
    Time Frame Starting from the first dose study treatment until the first documented CR or PR (every 8 weeks then after 8 cycles at every 12 weeks in duration of 94.0 weeks)

    Outcome Measure Data

    Analysis Population Description
    The response-evaluable population (RES) population included all enrolled participants who received at least 1 dose of study medication and had an adequate baseline tumor assessment.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 127
    Count of Participants [Participants]
    88
    69.3%
    2. Secondary Outcome
    Title IRR-Assessed Duration of Response (DR)
    Description DR: time from first documentation of objective tumor response (CR or PR) to first documentation of objective progressive disease (PD) or to death due to any cause, whichever occurred first. If no progression or death on study was observed, or given antitumor treatment other than study drug, participants were censored on date of last on-study tumor assessment. RECIST v1.1, a) PD: >=20% increase in sum of diameters of target lesions, taking as reference the smallest sum on study (this includes baseline sum if that is the smallest on study), sum must also demonstrate an absolute increase of >=5 mm, appearance of 1 or more new lesions, unequivocal progression of existing non-target lesions; b) CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); c) PR: >=30% decrease in sum of diameters of target lesions, taking as reference baseline sum diameters.
    Time Frame From first documentation of objective tumor response to first documentation of objective PD or death due to any cause, whichever occurred first (every 8 weeks then after 8 cycles at every 12 weeks in duration of 151.3 weeks)

    Outcome Measure Data

    Analysis Population Description
    The RES population included all enrolled participants who received at least 1 dose of study medication and had an adequate baseline tumor assessment. Here, "Overall Number of Participants Analyzed" signifies number of participants with a confirmed objective response that could have occurred anytime up to 151.3 weeks.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 91
    Median (95% Confidence Interval) [Months]
    19.7
    3. Secondary Outcome
    Title IRR-Assessed Time to Tumor Response (TTR)
    Description TTR was defined as the time from the date of first dose to first documentation of objective tumor response (CR or PR), that was subsequently confirmed. For participants proceeding from PR to CR, the onset of PR was taken as the onset of response. RECIST v1.1 (as determined by IRR), a) CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); b) PR: at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.
    Time Frame From date of first dose of crizotinib to first documentation of objective response was observed (every 8 weeks then after 8 cycles at every 12 weeks in duration of 151.3 weeks)

    Outcome Measure Data

    Analysis Population Description
    The RES population included all enrolled participants who received at least 1 dose of study medication and had an adequate baseline tumor assessment. Here, "Overall Number of Participants Analyzed" signifies number of participants with a confirmed objective response that could have occurred anytime up to 151.3 weeks.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 91
    Median (Full Range) [Months]
    1.9
    4. Secondary Outcome
    Title IRR Assessed Disease Control Rate (DCR) at 8 Weeks
    Description DCR at 8 weeks was defined as the percentage of participants with a confirmed CR, confirmed PR, or stable disease (SD) at 8 weeks, respectively, relative to the total population of response evaluable participants. RECIST v1.1 (as determined by IRR), a) CR: disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis); b) PR: at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study.
    Time Frame At 8 weeks after the start of study treatment

    Outcome Measure Data

    Analysis Population Description
    The RES population included all enrolled participants who received at least 1 dose of study medication and had an adequate baseline tumor assessment.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 127
    Number (95% Confidence Interval) [Percentage of participants]
    88.2
    69.4%
    5. Secondary Outcome
    Title IRR-Assessed Progression Free Survival (PFS)
    Description PFS was defined as the time from the date of the first dose of crizotinib to first documentation of objective PD or to death on study due to any cause, whichever occurred first. If no progression or death on study was observed, or given antitumor treatment other than study drug, participants were censored on date of last on-study tumor assessment. RECIST v1.1, PD: at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered a sign of progression. Unequivocal progression of existing non-target lesions.
    Time Frame From the date of first dose of crizotinib until the first documentation of objective PD or death (every 8 weeks then after 8 cycles at every 12 weeks in duration of 151.3 weeks)

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population (SAF) included all enrolled participants who received at least 1 dose of study medication.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 127
    Median (95% Confidence Interval) [Months]
    15.9
    6. Secondary Outcome
    Title Overall Survival (OS)
    Description OS was defined as the time from the date of the first dose of crizotinib to the date of death due to any cause. For participants still alive at the time of analysis, the OS time was censored on the last date the participants were known to be alive.
    Time Frame From date of the first dose of crizotinib until the date of death from any cause (up to 291.9 weeks)

    Outcome Measure Data

    Analysis Population Description
    SAF included all enrolled participants who received at least 1 dose of study medication.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 127
    Median (95% Confidence Interval) [Months]
    44.2
    7. Secondary Outcome
    Title Number of Participants With Treatment Emergent Adverse Events (AEs) and Serious AEs, Treatment Emergent Treatment Related AEs and SAEs, Grade 3 or 4 Treatment Emergent AEs and Grade 3 or 4 Treatment Emergent Treatment Related AEs
    Description Treatment-emergent AEs :between first dose of study drug and up to 28 days after last dose that were absent before treatment or that worsened relative to pretreatment state. Relatedness to crizotinib was assessed by the investigator. Treatment-related AE: any untoward medical occurrence attributed to study drug in a participant who received study drug. Serious adverse event (SAE):an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. Grade 3 (Severe) events=unacceptable or intolerable events, significantly interrupting usual daily activity, require systemic drug therapy/other treatment. Grade 4 (Life-threatening) events caused participant to be in imminent danger of death.
    Time Frame Baseline up to 28 days after the last dose of study treatment (maximum up to 295.9 weeks)

    Outcome Measure Data

    Analysis Population Description
    SAF included all enrolled participants who received at least 1 dose of study medication.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 127
    Incidence of All-Causality Adverse Events
    127
    100%
    Incidence of All-Causality Serious Adverse Events
    46
    36.2%
    Incidence of Treatment Related Adverse Events
    124
    97.6%
    Incidence of Treatment Related Serious Adverse Events
    11
    8.7%
    Incidence of All-Causality Grade 3-4 Adverse Event
    68
    53.5%
    Incidence of Treatment Related Grade 3-4 Adverse Event
    41
    32.3%
    8. Secondary Outcome
    Title Number of Participants With a Shift in Hematology Laboratory Results From Baseline Grade </=2 to Worst Grade 3 or Grade 4
    Description Laboratory values included hemoglobin increased, anemia, platelet count decreased, leukocytosis, white blood cell decreased, lymphocyte count increased, lymphocyte count decreased and neutrophil count decreased. Laboratory values were defined as National Cancer Institute Common Toxicity Criteria for Adverse Events Version 3.0 (NCI CTCAE v3.0) grade 3 or higher.
    Time Frame Baseline up to 28 days after the last dose of study treatment (maximum up to 295.9 weeks)

    Outcome Measure Data

    Analysis Population Description
    SAF included all enrolled participants who received at least 1 dose of study medication. Here, "number analyzed" signifies participants evaluable at specific rows.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 127
    Hemoglobin Increased: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Hemoglobin Increased: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hemoglobin Increased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Anemia: Baseline Grade 0 to Worst Grade 3
    2
    1.6%
    Anemia: Baseline Grade 1 to Worst Grade 3
    1
    0.8%
    Anemia: Baseline Grade 2 to Worst Grade 3
    5
    3.9%
    Anemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Anemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Anemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Platelet Count Decreased: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Platelet Count Decreased: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Platelet Count Decreased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Platelet Count Decreased: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Platelet Count Decreased: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Platelet Count Decreased: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Leukocytosis: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Leukocytosis: Baseline Grade 0 to Worst Grade 4
    0
    0%
    White Blood Cell Decreased: Baseline Grade 0 to Worst Grade 3
    3
    2.4%
    White Blood Cell Decreased: Baseline Grade 1 to Worst Grade 3
    1
    0.8%
    White Blood Cell Decreased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    White Blood Cell Decreased: Baseline Grade 0 to Worst Grade 4
    0
    0%
    White Blood Cell Decreased: Baseline Grade 1 to Worst Grade 4
    0
    0%
    White Blood Cell Decreased: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Lymphocyte Count Increased: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Lymphocyte Count Increased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Lymphocyte Count Decreased: Baseline Grade 0 to Worst Grade 3
    4
    3.1%
    Lymphocyte Count Decreased: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Lymphocyte Count Decreased: Baseline Grade 2 to Worst Grade 3
    2
    1.6%
    Lymphocyte Count Decreased: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Lymphocyte Count Decreased: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Lymphocyte Count Decreased: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Neutrophil Count Decreased: Baseline Grade 0 to Worst Grade 3
    7
    5.5%
    Neutrophil Count Decreased: Baseline Grade 1 to Worst Grade 3
    1
    0.8%
    Neutrophil Count Decreased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Neutrophil Count Decreased: Baseline Grade 0 to Worst Grade 4
    4
    3.1%
    Neutrophil Count Decreased: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Neutrophil Count Decreased: Baseline Grade 2 to Worst Grade 4
    0
    0%
    9. Secondary Outcome
    Title Number of Participants With a Shift of Chemistry Laboratory Results From Baseline Grade </=2 to Worst Grade 3 or Grade 4
    Description Laboratory values included blood bilirubin increased, alanine aminotransferase increased, aspartate aminotransferase increased, alkaline phosphatase increased, hypoalbuminemia, hypernatremia, hyponatremia, hyperkalemia, hypokalemia, hypercalcemia, hypocalcemia, hypophosphatemia, Creatinine increased, hyperuricemia, hypermagnesemia, hypomagnesemia, hyperglycemia and hypoglycemia. Laboratory values were defined as National Cancer Institute Common Toxicity Criteria for Adverse Events Version 3.0 (NCI CTCAE v3.0) grade 3 or higher.
    Time Frame Baseline up to 28 days after the last dose of study treatment (maximum up to 295.9 weeks)

    Outcome Measure Data

    Analysis Population Description
    SAF included all enrolled participants who received at least one dose of study medication. Here, "number analyzed" signifies participants evaluable at specific rows.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 127
    Blood Bilirubin Increased: Baseline Grade 0 to Worst Grade 3
    1
    0.8%
    Blood Bilirubin Increased: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Blood Bilirubin Increased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Blood Bilirubin Increased: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Blood Bilirubin Increased: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Blood Bilirubin Increased: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Alanine Aminotransferase Increased: Baseline Grade 0 to Worst Grade 3
    4
    3.1%
    Alanine Aminotransferase Increased: Baseline Grade 1 to Worst Grade 3
    1
    0.8%
    Alanine Aminotransferase Increased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Alanine Aminotransferase Increased: Baseline Grade 0 to Worst Grade 4
    4
    3.1%
    Alanine Aminotransferase Increased: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Alanine Aminotransferase Increased: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Aspartate Aminotransferase Increased : Baseline Grade 0 to Worst Grade 3
    2
    1.6%
    Aspartate Aminotransferase Increased : Baseline Grade 1 to Worst Grade 3
    0
    0%
    Aspartate Aminotransferase Increased : Baseline Grade 2 to Worst Grade 3
    0
    0%
    Aspartate Aminotransferase Increased : Baseline Grade 0 to Worst Grade 4
    4
    3.1%
    Aspartate Aminotransferase Increased : Baseline Grade 1 to Worst Grade 4
    0
    0%
    Aspartate Aminotransferase Increased : Baseline Grade 2 to Worst Grade 4
    0
    0%
    Alkaline Phosphatase Increased : Baseline Grade 0 to Worst Grade 3
    0
    0%
    Alkaline Phosphatase Increased : Baseline Grade 1 to Worst Grade 3
    0
    0%
    Alkaline Phosphatase Increased : Baseline Grade 2 to Worst Grade 3
    0
    0%
    Alkaline Phosphatase Increased : Baseline Grade 0 to Worst Grade 4
    0
    0%
    Alkaline Phosphatase Increased : Baseline Grade 1 to Worst Grade 4
    0
    0%
    Alkaline Phosphatase Increased : Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hypoalbuminemia: Baseline Grade 0 to Worst Grade 3
    1
    0.8%
    Hypoalbuminemia: Baseline Grade 1 to Worst Grade 3
    1
    0.8%
    Hypoalbuminemia: Baseline Grade 2 to Worst Grade 3
    1
    0.8%
    Hypoalbuminemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hypoalbuminemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypoalbuminemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hypernatremia: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Hypernatremia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypernatremia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hypernatremia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hypernatremia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypernatremia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hyponatremia: Baseline Grade 0 to Worst Grade 3
    8
    6.3%
    Hyponatremia: Baseline Grade 1 to Worst Grade 3
    4
    3.1%
    Hyponatremia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hyponatremia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hyperkalemia: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Hyperkalemia: Baseline Grade 1 to Worst Grade 3
    1
    0.8%
    Hyperkalemia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hyperkalemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hyperkalemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hyperkalemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hypokalemia: Baseline Grade 0 to Worst Grade 3
    5
    3.9%
    Hypokalemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypokalemia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hypokalemia: Baseline Grade 0 to Worst Grade 4
    1
    0.8%
    Hypokalemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypokalemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hypercalcemia: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Hypercalcemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypercalcemia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hypercalcemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hypercalcemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypercalcemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hypocalcemia: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Hypocalcemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypocalcemia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hypocalcemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hypocalcemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypocalcemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hypophosphatemia: Baseline Grade 0 to Worst Grade 3
    11
    8.7%
    Hypophosphatemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypophosphatemia: Baseline Grade 2 to Worst Grade 3
    1
    0.8%
    Hypophosphatemia: Baseline Grade 0 to Worst Grade 4
    1
    0.8%
    Hypophosphatemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypophosphatemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Creatinine Increased: Baseline Grade 0 to Worst Grade 3
    1
    0.8%
    Creatinine Increased: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Creatinine Increased: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Creatinine Increased: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Creatinine Increased: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Creatinine Increased: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hyperuricemia: Baseline Grade 0 to Worst Grade 3
    33
    26%
    Hyperuricemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hyperuricemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hyperuricemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypermagnesemia: Baseline Grade 0 to Worst Grade 3
    2
    1.6%
    Hypermagnesemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypermagnesemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hypermagnesemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypomagnesemia: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Hypomagnesemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypomagnesemia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hypomagnesemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hypomagnesemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypomagnesemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hyperglycemia: Baseline Grade 0 to Worst Grade 3
    1
    0.8%
    Hyperglycemia: Baseline Grade 1 to Worst Grade 3
    1
    0.8%
    Hyperglycemia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hyperglycemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hyperglycemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hyperglycemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    Hypoglycemia: Baseline Grade 0 to Worst Grade 3
    0
    0%
    Hypoglycemia: Baseline Grade 1 to Worst Grade 3
    0
    0%
    Hypoglycemia: Baseline Grade 2 to Worst Grade 3
    0
    0%
    Hypoglycemia: Baseline Grade 0 to Worst Grade 4
    0
    0%
    Hypoglycemia: Baseline Grade 1 to Worst Grade 4
    0
    0%
    Hypoglycemia: Baseline Grade 2 to Worst Grade 4
    0
    0%
    10. Secondary Outcome
    Title Change From Baseline to Cycle 60 in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life (QOL) Questionnaire Core 30 (QLQ-C30) Scores
    Description The EORTC QLQ-C30 consists of 30 questions which are incorporated into 5 functional domains (physical, role, cognitive, emotional, and social); a global health status/global QOL scale; 3 symptom scales (fatigue, pain, nausea and vomiting scales); and 6 single items that assess the additional symptoms (dyspnea, appetite loss, sleep disturbance/insomnia, constipation, and diarrhea) and the perceived financial burden of treatment. All the scales and single-item scores ranged from 0 to 100, higher score is indicative of a higher response level (high score for a functional scale represents a high / healthy level of functioning; high score for the global health status / QoL represents a high QoL; a high score for a symptom scale / item represents a high level of symptomatology / problems).
    Time Frame Baseline up to Cycle 60

    Outcome Measure Data

    Analysis Population Description
    Patient reported outcome (PRO)-evaluable population (PRO) included enrolled participants who received at least 1 dose of study medication and completed the assessments at baseline and with at least 1 post-baseline time point. Here, "Overall Number of Participants Analyzed" signifies participants evaluable for this outcome measure.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 26
    Global Health Status
    11.85
    (20.844)
    Physical Functioning
    3.08
    (11.964)
    Role Functioning
    -0.65
    (17.945)
    Emotional Functioning
    10.77
    (18.146)
    Cognitive Functioning
    3.20
    (14.159)
    Social Functioning
    5.13
    (20.961)
    Fatigue
    -12.30
    (20.156)
    Nausea and Vomiting
    0.64
    (14.517)
    Pain
    -12.18
    (16.027)
    Dyspnea
    -6.41
    (21.124)
    Insomnia
    -11.53
    (16.161)
    Appetite loss
    -14.10
    (23.428)
    Constipation
    10.26
    (27.920)
    Diarrhoea
    1.28
    (19.937)
    Financial difficulties
    -21.80
    (28.199)
    11. Secondary Outcome
    Title Change From Baseline to Cycle 60 in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Lung Cancer Module 13 Scores
    Description The EORTC QLQ-LC13 consisted of 1 multi-item scale and 9 single items that assessed specific symptoms (dyspnea, cough, hemoptysis, and site-specific pain), side effects (sore mouth, dysphagia, neuropathy, and alopecia). Scores on each scale and item ranged from 0 to 100, higher score is indicative of a higher response level (a high score for a symptom scale / item represents a high level of symptomatology / problems).
    Time Frame Baseline up to Cycle 60

    Outcome Measure Data

    Analysis Population Description
    PRO evaluable population included participants from the safety analysis population who completed the assessments at baseline and at least one post-baseline time point. Here, "Overall Number of Participants Analyzed" signifies participants evaluable for this outcome measure.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    Measure Participants 26
    Dyspnea
    -4.74
    (14.791)
    Coughing
    -12.82
    (29.936)
    Hemoptysis
    -5.12
    (12.253)
    Sore mouth
    -1.28
    (14.844)
    Dysphagia
    0.00
    (16.314)
    Peripheral neuropathy
    3.85
    (19.611)
    Alopecia
    -2.57
    (20.910)
    Pain in chest
    -12.82
    (21.236)
    Pain in arm or shoulder
    -15.37
    (23.536)
    Pain in other parts
    -7.68
    (21.718)

    Adverse Events

    Time Frame Baseline up to 28 days after the last dose of study treatment (maximum up to 295.9 weeks)
    Adverse Event Reporting Description Same event may appear as both an AE and SAE. However, what is presented are distinct events. An event may be categorized as serious in 1 participant and as non-serious in another, or a participant may have experienced both a serious and non-serious event.
    Arm/Group Title Crizotinib 250 mg
    Arm/Group Description Participants received crizotinib 250 mg orally twice a day in a cycle of 28 days. Dose was modified by investigator if there was treatment-related toxicity. Maximum treatment exposure time was of 291.9 weeks up to final analysis date.
    All Cause Mortality
    Crizotinib 250 mg
    Affected / at Risk (%) # Events
    Total 65/127 (51.2%)
    Serious Adverse Events
    Crizotinib 250 mg
    Affected / at Risk (%) # Events
    Total 46/127 (36.2%)
    Blood and lymphatic system disorders
    Anaemia 1/127 (0.8%)
    Cardiac disorders
    Acute myocardial infarction 1/127 (0.8%)
    Bradycardia 1/127 (0.8%)
    Eye disorders
    Retinal detachment 1/127 (0.8%)
    Gastrointestinal disorders
    Abdominal pain 1/127 (0.8%)
    Chronic gastritis 1/127 (0.8%)
    General disorders
    Disease Progression 7/127 (5.5%)
    Hernia 1/127 (0.8%)
    Peripheral swelling 1/127 (0.8%)
    Hepatobiliary disorders
    Hepatic cyst 1/127 (0.8%)
    Hepatic function abnormal 2/127 (1.6%)
    Drug-induced liver injury 1/127 (0.8%)
    Liver injury 1/127 (0.8%)
    Infections and infestations
    Pneumonia 10/127 (7.9%)
    Appendicitis 1/127 (0.8%)
    Bronchiolitis 1/127 (0.8%)
    Bronchopulmonary aspergillosis 1/127 (0.8%)
    Cellulitis 1/127 (0.8%)
    Empyema 1/127 (0.8%)
    Oesophageal infection 1/127 (0.8%)
    Urinary tract infection 1/127 (0.8%)
    Postoperative wound infection 1/127 (0.8%)
    Renal tuberculosis 1/127 (0.8%)
    Tuberculosis 1/127 (0.8%)
    Injury, poisoning and procedural complications
    Road traffic accident 1/127 (0.8%)
    Investigations
    Alanine aminotransferase increased 2/127 (1.6%)
    Aspartate aminotransferase increased 1/127 (0.8%)
    Hepatic enzyme increased 1/127 (0.8%)
    Metabolism and nutrition disorders
    Decreased appetite 1/127 (0.8%)
    Malnutrition 1/127 (0.8%)
    Hypoproteinaemia 1/127 (0.8%)
    Musculoskeletal and connective tissue disorders
    Spinal osteoarthritis 1/127 (0.8%)
    Arthropathy 1/127 (0.8%)
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Breast cancer 1/127 (0.8%)
    Colon cancer 1/127 (0.8%)
    Lung neoplasm malignant 1/127 (0.8%)
    Mediastinum neoplasm 1/127 (0.8%)
    Nervous system disorders
    Headache 1/127 (0.8%)
    Ruptured cerebral aneurysm 1/127 (0.8%)
    Seizure 1/127 (0.8%)
    Dysarthria 1/127 (0.8%)
    Epilepsy 1/127 (0.8%)
    Renal and urinary disorders
    Renal cyst 2/127 (1.6%)
    Chronic kidney disease 1/127 (0.8%)
    Respiratory, thoracic and mediastinal disorders
    Pleural effusion 3/127 (2.4%)
    Respiratory failure 5/127 (3.9%)
    Pneumothorax 1/127 (0.8%)
    Pulmonary embolism 1/127 (0.8%)
    Vascular disorders
    Deep vein thrombosis 2/127 (1.6%)
    Embolism 1/127 (0.8%)
    Other (Not Including Serious) Adverse Events
    Crizotinib 250 mg
    Affected / at Risk (%) # Events
    Total 127/127 (100%)
    Blood and lymphatic system disorders
    Neutropenia 14/127 (11%)
    Anaemia 15/127 (11.8%)
    Leukopenia 8/127 (6.3%)
    Cardiac disorders
    Sinus bradycardia 11/127 (8.7%)
    Eye disorders
    Vision blurred 25/127 (19.7%)
    Visual impairment 22/127 (17.3%)
    Diplopia 7/127 (5.5%)
    Photopsia 7/127 (5.5%)
    Gastrointestinal disorders
    Diarrhoea 62/127 (48.8%)
    Nausea 58/127 (45.7%)
    Vomiting 50/127 (39.4%)
    Constipation 49/127 (38.6%)
    Abdominal pain upper 12/127 (9.4%)
    Abdominal pain 12/127 (9.4%)
    Stomatitis 10/127 (7.9%)
    Abdominal distension 8/127 (6.3%)
    Toothache 10/127 (7.9%)
    General disorders
    Oedema peripheral 39/127 (30.7%)
    Fatigue 22/127 (17.3%)
    Pyrexia 23/127 (18.1%)
    Chest pain 11/127 (8.7%)
    Asthenia 9/127 (7.1%)
    Pain 9/127 (7.1%)
    Peripheral swelling 9/127 (7.1%)
    Hepatobiliary disorders
    Hepatic function abnormal 10/127 (7.9%)
    Infections and infestations
    Nasopharyngitis 28/127 (22%)
    Upper respiratory tract infection 22/127 (17.3%)
    Pneumonia 11/127 (8.7%)
    Urinary tract infection 11/127 (8.7%)
    Investigations
    Alanine aminotransferase increased 72/127 (56.7%)
    Aspartate aminotransferase increased 67/127 (52.8%)
    Neutrophil count decreased 37/127 (29.1%)
    White blood cell count decreased 30/127 (23.6%)
    Blood creatinine increased 29/127 (22.8%)
    Gamma-glutamyltransferase increased 18/127 (14.2%)
    Blood alkaline phosphate increased 14/127 (11%)
    Blood albumin decreased 17/127 (13.4%)
    Blood creatine phosphokinase increased 9/127 (7.1%)
    Protein total decreased 7/127 (5.5%)
    Weight decreased 8/127 (6.3%)
    Metabolism and nutrition disorders
    Decreased appetite 27/127 (21.3%)
    Hypoproteinaemia 14/127 (11%)
    Hypoalbuminaemia 18/127 (14.2%)
    Hypocalcaemia 10/127 (7.9%)
    Hypokalaemia 10/127 (7.9%)
    Hyponatraemia 12/127 (9.4%)
    Musculoskeletal and connective tissue disorders
    Back pain 21/127 (16.5%)
    Pain in extremity 16/127 (12.6%)
    Arthralgia 10/127 (7.9%)
    Musculoskeletal pain 10/127 (7.9%)
    Nervous system disorders
    Dysgeusia 17/127 (13.4%)
    Dizziness 23/127 (18.1%)
    Headache 20/127 (15.7%)
    Hypoaesthesia 7/127 (5.5%)
    Taste disorder 8/127 (6.3%)
    Psychiatric disorders
    Insomnia 10/127 (7.9%)
    Respiratory, thoracic and mediastinal disorders
    Cough 35/127 (27.6%)
    Dyspnoea 12/127 (9.4%)
    Haemoptysis 7/127 (5.5%)
    Skin and subcutaneous tissue disorders
    Rash 17/127 (13.4%)
    Pruritis 11/127 (8.7%)

    Limitations/Caveats

    Due to small percentage of participants (less than 20%) after Cycle 60, data for EORTC QLQ-C30 score and EORTC QLQ-LC13 score could not be interpreted. 65 participants died during the whole study: 15 participants died within 28 days from last dose of study treatment and were reported as SAEs, while 50 participants died during the survival follow-up.

    More Information

    Certain Agreements

    Principal Investigators are NOT employed by the organization sponsoring the study.

    Pfizer has the right to review disclosures, requesting a delay of less than 60 days. Investigator will postpone single center publications until after disclosure of pooled data (all sites), less than 12 months from study completion/termination at all participating sites. Investigator may not disclose previously undisclosed confidential information other than study results.

    Results Point of Contact

    Name/Title Pfizer ClinicalTrials.gov Call Center
    Organization Pfizer Inc.
    Phone 1-800-718-1021
    Email ClinicalTrials.gov_Inquiries@pfizer.com
    Responsible Party:
    Pfizer
    ClinicalTrials.gov Identifier:
    NCT01945021
    Other Study ID Numbers:
    • OO 12-01
    First Posted:
    Sep 18, 2013
    Last Update Posted:
    Feb 21, 2021
    Last Verified:
    Feb 1, 2021