A Study Of Lorlatinib Versus Crizotinib In First Line Treatment Of Patients With ALK-Positive NSCLC

Sponsor
Pfizer (Industry)
Overall Status
Active, not recruiting
CT.gov ID
NCT03052608
Collaborator
(none)
296
165
2
140.2
1.8
0

Study Details

Study Description

Brief Summary

A phase 3 study to demonstrate whether lorlatinib given as monotherapy is superior to crizotinib alone in prolonging the progression-free survival in advanced ALK-positive NSCLC patients who are treatment naïve and to compare lorlatinib to crizotinib with respect to overall survival in the same population

Condition or Disease Intervention/Treatment Phase
Phase 3

Study Design

Study Type:
Interventional
Actual Enrollment :
296 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
A PHASE 3, RANDOMIZED, OPEN-LABEL STUDY OF LORLATINIB (PF-06463922) MONOTHERAPY VERSUS CRIZOTINIB MONOTHERAPY IN THE FIRST-LINE TREATMENT OF PATIENTS WITH ADVANCED ALK-POSITIVE NON-SMALL CELL LUNG CANCER
Actual Study Start Date :
Apr 27, 2017
Actual Primary Completion Date :
Mar 20, 2020
Anticipated Study Completion Date :
Dec 31, 2028

Arms and Interventions

Arm Intervention/Treatment
Experimental: Lorlatinib

Lorlatinib single agent, 100 mg (4 x 25 mg) oral tables, QD, continuously

Drug: Lorlatinib
ALK-positive NSCL treatment
Other Names:
  • PF-06463922
  • Active Comparator: Crizotinib

    Crizotinib single agent, 250 mg (1 x 250) oral capsules, BID, continuously

    Drug: Crizotinib
    ALK-positive NSCL treatment
    Other Names:
  • Xalkori
  • Outcome Measures

    Primary Outcome Measures

    1. Progression-Free Survival (PFS) Based on Blinded Independent Central Review (BICR) Assessment [From time of Study Start up to 33 months]

      PFS was defined as the time from randomization to the date of the first documentation of progressive disease as assessed by the independent radiologist or death due to any cause, whichever occurred first. PFS (in months) was calculated as (date of event or censoring-randomization+1)/30.4375. Progressive disease is defined per RECIST version 1.1, as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.

    Secondary Outcome Measures

    1. Overall Survival (OS) [From time of Study Start up to 33 months]

      OS was defined as the time from randomization to the date of death due to any cause. OS (in months) was calculated as (date of death or censoring - start date +1)/30.4375. Participants last known to be alive were censored at date of last contact.

    2. Progression-Free Survival (PFS) Based on Investigator's Assessment [From time of Study Start up to 33 months]

      PFS was defined as the time from randomization to the date of the first documentation of progressive disease as assessed by investigator or death due to any cause, whichever occurred first. PFS (in months) was calculated as (date of event or censoring-randomization+1)/30.4375. Progressive disease is defined per RECIST version 1.1, as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.

    3. Objective Response Rate (ORR) - Percentage of Participants With Objective Response (OR) Based on BICR Assessment [From time of Study Start up to 33 months]

      ORR was the percentage of participants with objective response of complete response (CR) or partial response (PR) according to RECIST version 1.1 recorded from randomization until disease progression or start of new anti-cancer therapy. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.

    4. Objective Response Rate (ORR) - Percentage of Participants With Objective Response (OR) Based on Investigator's Assessment [From time of Study Start up to 33 months]

      ORR was the percentage of participants with objective response of complete response (CR) or partial response (PR) according to RECIST version 1.1 recorded from randomization until disease progression or start of new anti-cancer therapy. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.

    5. Intracranial Objective Response Rate (IC-ORR) - Percentage of Participants With Intracranial Objective Response (IC-OR) Based on BICR Assessment [From time of Study Start up to 33 months]

      IC-ORR was the percentage of participant with intracranial objective response of complete response (CR) or partial response (PR) based on intracranial disease in the subset of participants with at least 1 intracranial lesion per RECIST version 1.1 (modified) recorded from randomization until disease progression or start of new anti-cancer therapy. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.

    6. Intracranial Time to Progression (IC-TTP) Based on BICR Assessment [From time of Study Start up to 33 months]

      IC-TTP based on BICR assessment was defined as the time from date of randomization to the date of the first documentation of progression of intracranial disease, based on either new brain metastases or progression of existing brain metastases.

    7. Duration of Response (DR) Based on BICR Assessment [From time of Study Start up to 33 months]

      DR was defined, for participants with a confirmed objective response (OR) of complete response (CR) or partial response (PR) per RECIST version 1.1, as the time from the first documentation of OR to the first documentation of progressive disease (PD) or death due to any cause, whichever occurred first. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions. PD was defined as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.

    8. Intracranial Duration of Response (IC-DR) Based on BICR Assessment [From time of Study Start up to 33 months]

      IC-DR was defined, for participants with a confirmed intracranial objective response (OR) of complete response (CR) or partial response (PR) per RECIST version 1.1, as the time from the first documentation of intracranial OR to the first documentation of intracranial progressive disease (PD) or death due to any cause, whichever occurred first. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions. PD was defined as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.

    9. Time to Tumor Response (TTR) Based on BICR Assessment [From time of Study Start up to 33 months]

      TTR based on BICR assessment was defined, for participants with a confirmed objective response, as the time from the date of randomization to the first documentation of objective response (complete response or partial response) which was subsequently confirmed. Complete response was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). Partial response was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.

    10. Intracranial Time to Tumor Response (IC-TTR) Based on BICR Assessment [From time of Study Start up to 33 months]

      IC-TTR was defined, for participants with a confirmed intracranial objective response, as the time from the date of randomization to the first documentation of intracranial objective response (complete response or partial response) which was subsequently confirmed. Complete response was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). Partial response was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.

    11. PFS2 Based on Investigator's Assessment [From time of Study Start up to 45 months]

      PFS2 was defined as the time from randomization to the date of progression of disease on first subsequent systemic anti-cancer therapy, or death from any cause, whichever occurred first

    12. Number of Participants With Treatment-Emergent Adverse Events (AEs; All-Causality and Treatment-Related) [From time of Study Start up to 33 months]

      An AE was an untoward medical occurrence in a participant who received study treatment without regard to possibility of causal relationship. Serious adverse event (SAE) was an AE resulting in any of the following outcomes: death, life-threatening experience, initial or prolonged inpatient hospitalization, persistent or significant disability/incapacity, congenital anomaly/birth defect. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. All AEs in the table below were treatment-emergent AEs. Grade 3 and 4 AEs in the table below indicated severe AE and life-threatening consequences respectively; Grade 5 indicated death due to AE. Treatment-related AEs were determined by investigators.

    13. Number of Participants With Changes in Hematology Laboratory Parameters From Baseline Maximum NCI-CTCAE Grade Lower Than 3 to Postbaseline Maximum Grade 3 or Grade 4 [From Baseline up to 33 months]

      Laboratory test results were graded according to National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.03. The CTCAE displays Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grades 1 and 2 indicate mild and moderate AE, respectively; Grades 3 and 4 indicate severe AE and life-threatening consequences respectively; Grade 5 indicates death due to AE. Participants with laboratory test abnormalities were summarized according to the worst grade for each laboratory test result. All participants meeting the postbaseline grade criteria in the table below had baseline maximum grades lower than 3.

    14. Number of Participants With Changes in Chemistry Laboratory Parameters From Baseline Maximum NCI-CTCAE Grade Lower Than 3 to Postbaseline Maximum Grade 3 or Grade 4 [From Baseline up to 33 months]

      Laboratory test results were graded according to National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.03. The CTCAE displays Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grades 1 and 2 indicate mild and moderate AE, respectively; Grades 3 and 4 indicate severe AE and life-threatening consequences respectively; Grade 5 indicates death due to AE. Participants with laboratory test abnormalities were summarized according to the worst grade for each laboratory test result. All participants meeting the postbaseline grade criteria in the table below had baseline maximum grades lower than 3.

    15. Number of Participants With Changes in Lipid Laboratory Parameters From Baseline Maximum NCI-CTCAE Grade Lower Than 3 to Postbaseline Maximum Grade 3 or Grade 4 [From Baseline up to 33 months]

      Laboratory test results were graded according to National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.03. The CTCAE displays Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grades 1 and 2 indicate mild and moderate AE, respectively; Grades 3 and 4 indicate severe AE and life-threatening consequences respectively; Grade 5 indicates death due to AE. Participants with laboratory test abnormalities were summarized according to the worst grade for each laboratory test result. All participants meeting the postbaseline grade criteria in the table below had baseline maximum grades lower than 3.

    16. Number of Participant With Vital Signs and Body Weight Data Meeting Pre-defined Criteria [From Baseline up to 33 months]

      Vital signs data included pulse, systolic blood pressure, and diastolic blood pressure. Measurements were only provided once per timepoint. If multiple assessments were provided per timepoint, the maximum value were used for reporting. The pre-defined criteria of vital sign and body weight data were as follows: maximum pulse rate >120 beats per minute (bpm); minimum pulse rate <50 bpm; maximum increase in pulse rate ≥30 bpm; maximum decrease in pulse rate ≥30 bpm; increase in systolic blood Pressure ≥40 mmHg; decrease in systolic blood pressure ≥40 mmHg; decrease in systolic blood pressure ≥60 mmHg; increase in diastolic blood pressure ≥20 mmHg; decrease in diastolic blood pressure ≥20 mmHg; decrease in diastolic blood pressure ≥40 mmHg; increase in body weight ≥10%; increase in body weight ≥20%; decrease in body weight ≥10%.

    17. Number of Participant With Maximum Increase From Baseline in Electrocardiogram (ECG) Data Meeting Pre-defined Criteria [From Baseline up to 33 months]

      Baseline was defined as the last assessment performed on or prior to date of the first dose of study treatment. Triplicate ECGs were collected in the study and the average of the replicate assessments were used for summary analysis. The pre-defined criteria of ECG data were as follows: change from baseline in QTcF ≥60 msec, ≥30 msec but <60 msec, <30 msec; change from baseline in QTcB ≥60 msec, ≥30 msec but <60 msec, <30 msec; PR change ≥50% if absolute baseline value was <200 msec; PR change ≥25% if absolute baseline value was ≥200 msec; QRS change ≥50% if absolute baseline value was <100 msec; QRS change ≥25% if absolute baseline value was ≥100 msec.

    18. Number of Participants With Maximum Decrease From Baseline Greater Than or Equal to 20 Points in Left Ventricular Ejection Fraction (LVEF) Percentage [From Baseline up to 33 months]

      In this outcome measure, baseline was defined as the last assessment on or prior to the date of the first dose of study treatment. Decrease from baseline was an absolute difference between baseline and observed value.

    19. Change From Baseline in Total Scores of Beck Depression Inventory (BDI)-II (Mood Assessment) Across Time [Baseline, Cycle 2 Day 1 (C2D1), C3D1, C4D1, C5D1, C6D1, C8D1, C10D1, C12D1, C14D1, C16D1, C18D1, C20D1, C22D1, C24D1, C26D1, C28D1, C30D1, C32D1, C34D1, C36D1, C38D1 and End of Treatment]

      BDI-II (Mood Assessment) is a 21 item self-reported scale, with each item rated by participants on a 4-point scale (ranging from 0-3). The scale includes items capturing mood, (loss of pleasure, sadness, and irritability), suicidal ideation, and cognitive signs (punitive thoughts, self-criticism, self-dislike pessimism, poor concentration) as well as somatic signs (appetite, sleep, fatigue, libido). Scores were obtained by adding up the total points from the series of answers. The total score ranged from 0 to 63, with higher total scores indicating more severe depressive symptoms. The standardized cutoffs are as follows: 0-13: minimal depression; 14-19: mild depression; 20-28: moderate depression; 29-63: severe depression.

    20. Number of Participants With Suicidal Ideation and Suicidal Behavior Across Time [Baseline, Cycle 2 Day 1 (C2D1), C3D1, C4D1, C5D1, C6D1, C8D1, C10D1, C12D1, C14D1, C16D1, C18D1, C20D1, C22D1, C24D1, C26D1, C28D1, C30D1, C32D1, C34D1, C36D1, C38D1 and End of Treatment]

      Suicidal ideation and behaviors were assessed by the Columbia Suicide Severity Rating Scale (C-SSRS). The C-SSRS is a unique, simple and short method of assessing both behavior and ideation that tracks all suicidal events and provides a summary of suicidality. It assesses the lethality of attempts and other features of ideation (frequency, duration, controllability, reasons for ideation, and deterrents), all of which are significantly predictive of completed suicide.

    21. Change From Baseline in Global Quality of Life (QOL), Functional Scales and Symptoms Scales as Assessed by the European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC-QLQ-C30) During Overall Treatment [From Baseline up to Cycle 38 Day 1]

      The EORTC QLQ C30 consists of 30 questions and includes 5 functional scales (physical, role, cognitive, emotional, and social); a global health status/global quality of life scale; 3 symptom scales (fatigue, pain, nausea and vomiting); and 6 single items that assess additional symptoms (dyspnea, appetite loss, sleep disturbance, constipation, and diarrhea) and financial impact. All scales and single item measures range in score from 0 to 100. Higher scores on the functional scales represent higher levels of functioning. Higher scores on the global health status/quality of life scale represent higher health status/quality of life. Higher scores on symptom scales/items represent a greater presence of symptoms.

    22. Change From Baseline in Lung Cancer Symptoms as Assessed by the EORTC Quality of Life Questionnaire-Lung Cancer 13 (QLQ- LC13) During Overall Treatment [From Baseline up to Cycle 38 Day 1]

      The EORTC QLQ LC13 consists of 13 questions and includes 1 multi-item scale and 9 single items assessing symptoms (dyspnea, cough, haemoptysis, and site-specific pain), side effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication use. The scale scores rang from 0 to 100, with higher scores indicating higher ("worse") level of symptoms.

    23. Change From Baseline in Health Status as Assessed by EuroQol 5 Dimension 5 Level (EQ-5D-5L) - Visual Analogue Scale (VAS) Across Time [Baseline, Day 1 of all cycles from Cycle 2 to Cycle 38, and End of Treatment]

      The EuroQol EQ-5D-5L is a participant-completed questionnaire designed to assess health status. There are 2 components to the EuroQol EQ-5D-5L: a descriptive system in which individuals rate their level of problems (none, slight, moderate, severe, extreme/unable) in 5 areas (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) and a VAS. The VAS component rates current health state on a scale from 0 (worst imaginable health state) to 100 (best imaginable health state) and higher scores indicate better health state.

    24. Change From Baseline in Health Status as Assessed by EuroQol 5 Dimension 5 Level (EQ-5D-5L) - Index Across Time [Baseline, Day 1 of all cycles from Cycle 2 to Cycle 38, and End of Treatment]

      The EuroQol EQ-5D-5L is a participant-completed questionnaire designed to assess health status. There are 2 components to the EuroQol EQ-5D-5L: a descriptive system in which individuals rate their level of problems (none, slight, moderate, severe, extreme/unable) in 5 areas (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) and a VAS in which participants rate their overall health status from 0 (worst imaginable) to 100 (best imaginable). EQ-5D summary index is obtained with a formula that weights each level of the 5 dimensions. The index-based score is interpreted along a continuum of 0 (death) to 1 (perfect health).

    25. Time to Deterioration (TTD) in Participant Reported Pain in Chest, Dyspnea, or Cough From QLQ-LC13 [From Baseline up to 33 months]

      The EORTC QLQ LC13 consists of 13 questions and includes 1 multi-item scale and 9 single items assessing symptoms (dyspnea, cough, haemoptysis, and site-specific pain), side effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication use. The scale scores rang from 0 to 100, with higher scores indicating higher ("worse") level of symptoms. TTD in pain in chest, dyspnea, or cough was defined as the time from randomization to the first time the participant's score showed a 10 point or greater increase after baseline in any of the 3 symptoms. TTD in months was calculated as (date of deterioration or censoring - randomization date +1)/30.4375.

    26. Number of Participant With ALK Domain Mutation in Plasma Circulating Nucleic Acid (CNA) Analysis at Screening, Cycle 2 Day 1 and Cycle 7 Day 1 [at Screening, Cycle 2 Day 1 and Cycle 7 Day 1]

      The analysis of anaplastic lymphoma kinase (ALK) domain mutation in plasma CNA was performed by next-generation sequencing (NGS) and the number of participants with one or more ALK mutations at Screening, Cycle 2 Day 1 and Cycle 7 Day 1 is presented here.

    27. Number of Participant With ALK Fusion Variant in Plasma Circulating Nucleic Acid (CNA) Analysis at Screening, Cycle 2 Day 1 and Cycle 7 Day 1 [at Screening, Cycle 2 Day 1 and Cycle 7 Day 1]

      The analysis of ALK fusion variant in plasma CNA was performed by NGS and the number of participants with fusion variants at Screening, Cycle 2 Day 1 and Cycle 7 Day 1 is presented here. In the table below, EML4-ALK is the abbreviation of echinoderm microtubule-associated protein-like 4 anaplastic lymphoma kinase.

    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 confirmed diagnosis of locally advanced or metastatic ALK-positive NSCLC; at least 1 extracranial measurable target lesion not previously irradiated. CNS metastases allowed if asymptomatic and not currently requiring corticosteroid treatment.

    • Availability of an archival FFPE tissue specimen.

    • No prior systemic NSCLC treatment.

    • ECOG PS 0, 1, or 2.

    • Age ≥18 years .

    • Adequate Bone Marrow, Liver, Renal, Pancreatic Function

    • Negative pregnancy test for females of childbearing potential

    Exclusion Criteria:
    • Spinal cord compression unless good pain control attained

    • Major surgery within 4 weeks prior to randomization.

    • Radiation therapy within 2 weeks prior to randomization, including stereotactic or partial brain irradiation. Whole brain irradiation within 4 weeks prior to randomization

    • Active bacterial, fungal, or viral infection

    • Clinically significant cardiovascular disease, active or within 3 months prior to enrollment. Ongoing cardiac dysrhythmias, uncontrolled atrial fibrillation, bradycardia or congenital long QT syndrome

    • Predisposing characteristics for acute pancreatitis in the last month prior to randomization.

    • History of extensive, disseminated, bilateral or presence of Grade 3 or 4 interstitial fibrosis or interstitial lung disease

    • Active malignancy (other than NSCLC, non melanoma skin cancer, in situ cervical cancer, papillary thyroid cancer, LCIS/DCIS of the breast, or localized prostate cancer) within the last 3 years prior to randomization.

    • Concurrent use of any of the following food or drugs within 12 days prior to the first dose of lorlatinib or crizotinib.

    1. known strong CYP3A inhibitors .

    2. known strong CYP3A inducers

    3. known P gp substrates with a narrow therapeutic index

    • Concurrent use of CYP3A substrates with narrow therapeutic indices within 12 days prior to the first dose of lorlatinib or crizotinib.

    • Other severe acute or chronic medical or psychiatric condition, including recent or active suicidal ideation or behavior, or laboratory abnormality that may increase the risk associated with study participation or interfere with the interpretation of study results

    • Investigational site staff members directly involved in the conduct of the study and their family members, or Pfizer employees, including their family members, directly involved in the conduct of the study.

    • Participation in other studies involving investigational drug(s) within 2 weeks prior to study entry and/or during study participation.

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Florida Cancer Specialists Altamonte Springs Florida United States 32701
    2 Florida Cancer Specialists Brandon Florida United States 33511
    3 Florida Cancer Specialists Clearwater Florida United States 33761
    4 Florida Cancer Specialists Gainesville Florida United States 32605
    5 Florida Cancer Specialists Largo Florida United States 33770
    6 Florida Cancer Specialists Lecanto Florida United States 34461
    7 Florida Cancer Specialists Orange City Florida United States 32763
    8 Florida Cancer Specialists Orlando Florida United States 32806
    9 Florida Cancer Specialists Saint Petersburg Florida United States 33705
    10 Florida Cancer Specialists Spring Hill Florida United States 34608
    11 Florida Cancer Specialists Tampa Florida United States 33607
    12 Florida Cancer Specialists Tavares Florida United States 32778
    13 Florida Cancer Specialists The Villages Florida United States 32159
    14 Florida Cancer Specialists Winter Park Florida United States 32792
    15 Massachusetts Eye and Ear Infirmary Boston Massachusetts United States 02114
    16 Massachusetts General Hospital Boston Massachusetts United States 02114
    17 Ophthalmic Consultants of Boston Inc Boston Massachusetts United States 02114
    18 The William P. Beetham Eye Institute, Joslin Diabetes Center Boston Massachusetts United States 02215
    19 University of Rochester Cancer Center Pharmacy Rochester New York United States 14642
    20 University of Rochester Medical Center Rochester New York United States 14642
    21 Tennessee Oncology, PLLC Dickson Tennessee United States 37055
    22 Tennessee Oncology, PLLC Franklin Tennessee United States 37067
    23 Tennessee Oncology, PLLC Gallatin Tennessee United States 37066
    24 Tennessee Oncology, PLLC Hermitage Tennessee United States 37076
    25 Tennessee Oncology, PLLC Lebanon Tennessee United States 37090
    26 Tennessee Oncology, PLLC Murfreesboro Tennessee United States 37129
    27 Tennessee Oncology, PLLC Nashville Tennessee United States 37203
    28 The Sarah Cannon Research Institute. Nashville Tennessee United States 37203
    29 Tennessee Oncology, PLLC Nashville Tennessee United States 37205
    30 Tennessee Oncology, PLLC Nashville Tennessee United States 37207
    31 Tennessee Oncology, PLLC Nashville Tennessee United States 37211
    32 Tennessee Oncology, PLLC Shelbyville Tennessee United States 37160
    33 Tennessee Oncology, PLLC Smyrna Tennessee United States 37167
    34 University of Washington Medical Center Seattle Washington United States 98195
    35 Centro de Investigacion Pergamino SA Pergamino Buenos Aires Argentina B2700CPM
    36 Centro Medico Austral Caba Argentina C1019ABS
    37 Bendigo Day Surgery Collection Centre and Laboratory Bendigo Victoria Australia 3550
    38 Bendigo Medical Imaging, Bendigo Hospital Bendigo Victoria Australia 3550
    39 Melbourne Pathology Bendigo Victoria Australia 3550
    40 Peter MacCallum Cancer Centre Melbourne Victoria Australia 3000
    41 Grand Hopital de Charleroi - Site Notre Dame Charleroi Belgium 6000
    42 Princess Margaret Cancer Centre Toronto Ontario Canada M5G 2M9
    43 McGill University Health Centre Montreal Quebec Canada H4A 3J1
    44 The Affiliated Hospital of Military Medical Sciences Beijing Beijing China 100071
    45 Jilin Provincial Cancer Hospital Changchun Jilin China 130012
    46 Shanghai Chest Hospital Shanghai Shanghai China 200030
    47 Department of Respiratory,the First Affiliated Hospital of College of Medicine, Zhejiang University Hangzhou Zhejiang China 310003
    48 The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou Zhejiang China 310009
    49 Zhejiang Cancer Hospital Hangzhou Zhejiang China 310022
    50 Beijing Cancer Hospital Beijing China 100142
    51 Guangdong General Hospital Guangzhou China 510000
    52 Fakultni nemocnice Olomouc, Klinika plicnich nemoci a tuberkulozy Olomouc Czechia 779 00
    53 Vseobecna fakultni nemocnice v Praze Praha 2 Czechia 128 08
    54 Hopital Haut-Léveque-Centre François Magendie Pessac Aquitaine France 33604
    55 Hopital de Chevilly Larue Chevilly Larue France 94550
    56 Centre Hospitalier du Mans Le Mans France 72000
    57 Institut Paoli-Calmettes Marseille cedex 09 France 13273
    58 Département d'Imagerie Médicale Marseille cedex 20 France 13915
    59 Hôpital Nord Marseille cedex 20 France 13915
    60 Service Ophtalmologie Marseille cedex 20 France 13915
    61 Groupe Hospitalier Bichat Claude Bernard, AP-HP Paris France 75018
    62 CHU de Rennes Hopital Pontchaillou Rennes cedex 9 France 35033
    63 CHU de Rennes, Hopital Pontchaillou Rennes cedex 9 France 35033
    64 Hopital Foch Suresnes France 92150
    65 Hopital Larrey Toulouse Cedex 9 France 31059
    66 Hopital Pierre Paul Riquet Toulouse cedex 9 France 31059
    67 Hôpital Larrey Toulouse Cedex 9 France 31059
    68 Department d'imagerie medicale Villejuif France 94805
    69 Institut Gustave Roussy Villejuif France 94805
    70 Uberortliche Radiologische Gemeinschaftspraxis Dr. med. Marc Amler Dresden Germany 01309
    71 Technische Universitat Dresden , Medizinische Fakultat Carl Gustav Carus Dresden Germany D-01307
    72 Thoraxklinik Heidelberg gGmbH Heidelberg Germany D-69126
    73 Universitatsklinikum Heidelberg Heidelberg Germany D-69126
    74 Universitätsklinikum des Saarlandes, Innere Medizin V Homburg - Saar Germany 66421
    75 Universitätsklinikum des Saarlandes Homburg - Saar Germany 66421
    76 Universitatsklinikum Regensburg, Institut fur Rontgendiagnostik Regensburg Germany 93053
    77 Universitatsklinikum Regensburg, Klinik und Poliklinik fur Innere Medizin II Regensburg Germany 93053
    78 The University of Hong Kong, Department of Clinical Oncology Hong Kong Hong Kong
    79 The University of Hong Kong, Department of Medicine Hong Kong Hong Kong
    80 Tuen Mun Hospital Hong Kong Hong Kong
    81 Artemis Hospital Gurugram Haryana India 122001
    82 Manipal Hospital Bangalore Karnataka India 560017
    83 Srinivasam Cancer Care Multispeciality Hospitals India Pvt Ltd Bangalore Karnataka India 560072
    84 Sahyadri Clinical Research and Development Centre Pune Maharashtra India 411004
    85 Sahyadri Specialty Hospital Pune Maharashtra India 411004
    86 AOU Policlinico Vittorio Emanuele-P.O.G. Rodolico Catania CT Italy 95123
    87 ASST Monza - A.O. San Gerardo Monza MB Italy 20900
    88 IRCCS Ospedale San Raffaele Milano MI Italy 20132
    89 Fondazione IRCCS Istituto Nazionale dei Tumori Milano MI Italy 20133
    90 Istituto Europeo di Oncologia Milano MI Italy 20141
    91 Istituto Clinico Humanitas Rozzano MI Italy 20089
    92 Azienda Ospedaliera di Perugia - Ospedale S. M. Misericordia Perugia PG Italy 06132
    93 Centro di Riferimento Oncologico-IRCCS Pordenone PN Italy 33081
    94 Azienda Ospedaliero-Universitaria di Parma Parma PR Italy 43126
    95 Istituto Nazionale Tumori Regina Elena Roma RM Italy 00144
    96 Az.Osp.San Camillo-Forlanini Roma RM Italy 00152
    97 Azienda Ospedaliera Dei Colli Ospedale Monaldi Napoli Italy 80131
    98 Istituto Nazionale Tumori di Napoli Napoli Italy 80131
    99 Ausl della Romagna- Ravenna Ravenna Italy 48121
    100 Aichi cancer center central hospital Nagoya Aichi Japan 464-8681
    101 Nagoya University Hospital Nagoya Aichi Japan 466-8560
    102 National Hospital Organization Shikoku Cancer Center Matsuyama Ehime Japan 791-0280
    103 Kurume University Hospital Kurume Fukuoka Japan 830-0011
    104 National Hospital Organization Hokkaido Cancer Center Sapporo Hokkaido Japan 003-0804
    105 Kanazawa University Hospital Kanazawa Ishikawa Japan 920-8641
    106 Kanagawa Cancer Center Yokohama Kanagawa Japan 241-8515
    107 Sendai Kousei Hospital Sendai Miyagi Japan 980-0873
    108 Osaka International Cancer Institute Osaka-shi Osaka Japan 541-8567
    109 Kindai University Hospital Osakasayama Osaka Japan 589-8511
    110 Shizuoka Cancer Center Sunto-gun Shizuoka Japan 411-8777
    111 National Cancer Center Hospital Chuo-Ku Tokyo Japan 104-0045
    112 National Hospital Organization, Yamaguchi-Ube Medical Center Ube Yamaguchi Japan 755-0241
    113 The Cancer Institute Hospital of JFCR Koto-ku, Tokyo Japan 135-8550
    114 Niigata Cancer Center Hospital Niigata Japan 951-8566
    115 Okayama University Hospital Okayama Japan 700-8558
    116 Osaka City General Hospital Osaka Japan 534-0021
    117 Tokushima University Hospital Tokushima Japan 770-8503
    118 Wakayama Medical University Department of Pulmonary Medicine and Oncology Wakayama Japan 641-8509
    119 The Catholic University of Korea, St. Vincents Hospital Suwon Gyeonggi-do Korea, Republic of 16247
    120 Seoul National University Hospital Seoul Korea, Republic of 03080
    121 Division of Medical Oncology, Severance Hospital, Yonsei University Health System Seoul Korea, Republic of 03722
    122 Asan Medical Center Seoul Korea, Republic of 05505
    123 Ulsan University Hospital Ulsan Korea, Republic of 44033
    124 Médicos Especialistas en Cancer S.C. / San Peregrino. Aguascalientes Mexico 20230
    125 Instituto Nacional de Cancerologia Distrito Federal Mexico 14080
    126 Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas Distrito Federal Mexico 14080
    127 University Medical Center Groningen Groningen Netherlands 9713 GZ
    128 Klinika Onkologii i Radioterapii Uniwersyteckie Centrum Kliniczne Gdansk Poland 80-214
    129 Med-Polonia Sp. z o.o. Poznan Poland 60-693
    130 Centrum Medyczne Dom Lekarski S.A. Szczecin Poland 70-784
    131 Narodowy Instytut Onkologii im. Marii Sklodowskiej-Curie-Panstwowy Instytut Badawczy Warszawa Poland 02-781
    132 RBHI Kursk Regional Clinical Oncology Dispensary Kislino, Ryshkovsky Rural Council Kursk Region Russian Federation 305 524
    133 Budgetary Healthcare Institution Omsk Region "Clinical Oncological Dispensary" Omsk OMSK Region Russian Federation 644013
    134 LEC at SBIH "Saint-Petersburg Clinical Research Practical Center of specialized types of Pesochniy Poselok Saint-petersburg Russian Federation 197758
    135 Private Medical Institution "Euromedservice" Pushkin Saint-petersburg Russian Federation 196603
    136 RBHI Kursk Regional Clinical Oncology Dispensary Kursk Russian Federation 305 035
    137 FSBI "N.N.Blokhin Medical Research Center of Oncology" Moscow Russian Federation 115478
    138 National University Hospital Singapore Singapore 119074
    139 Raffles Hospital Singapore Singapore 188770
    140 Institut Catala d'Oncologia L'Hospitalet L'Hospitalet de Llobregat Barcelona Spain 08908
    141 Hospital Universitario Insular de Gran Canaria Las Palmas de Gran Canaria Canarias Spain 35016
    142 Hospital Universitario Puerta de Hierro Majadahonda Majadahonda Madrid Spain 28222
    143 Complejo Hospitalario De Navarra Pamplona Navarra Spain 31008
    144 Hospital Teresa Herrera C.H.U.A.C. A Coruna Spain 15006
    145 Hospital De La Santa Creu I Sant Pau Barcelona Spain 08025
    146 Hospital Universitari Vall d'Hebron Barcelona Spain 08035
    147 Hospital Clinic i Provincial de Barcelona Barcelona Spain 08036
    148 Hospital De La Santa Creu I Sant Pau Barcelona Spain 08041
    149 Institut Catala d'Oncologia Girona Girona Spain 17007
    150 Hospital Universitario Ramon y Cajal Madrid Spain 28034
    151 Hospital Clinico San Carlos Madrid Spain 28040
    152 Hospital Universitario Fundacion Jimenez Diaz Madrid Spain 28040
    153 Hospital Universitario La Fe Valencia Spain 46026
    154 National Taiwan University Hospital Taipei Taiwan ROC Taiwan 10002
    155 Chang Gung Memorial Hospital - Kaohsiung Branch Kaohsiung Taiwan 83301
    156 Chung Shan Medical University Hospital Taichung Taiwan 402
    157 Taichung Veterans General Hospital Taichung Taiwan 40705
    158 Taipei Veterans General Hospital Taipei Taiwan 112
    159 Cukurova University Medical Faculty Adana Turkey 01330
    160 Istanbul University Oncology Institute Istanbul Turkey 34093
    161 Marmara Univ Pendik Training and Research Hospital Istanbul Turkey 34899
    162 Ege University Medical Faculty Izmir Turkey 35100
    163 The Ipswich Hospital NHS Trust Ipswich Suffolk United Kingdom IP4 5PD
    164 Heart of England NHS Foundation Trust, Birmingham Heartlands Hospital Birmingham WEST Midlands United Kingdom B9 5SS
    165 The Christie NHS Foundation Trust Manchester United Kingdom M20 4BX

    Sponsors and Collaborators

    • Pfizer

    Investigators

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

    Study Documents (Full-Text)

    More Information

    Additional Information:

    Publications

    None provided.
    Responsible Party:
    Pfizer
    ClinicalTrials.gov Identifier:
    NCT03052608
    Other Study ID Numbers:
    • B7461006
    • 2016-003315-35
    First Posted:
    Feb 14, 2017
    Last Update Posted:
    May 31, 2022
    Last Verified:
    May 1, 2022
    Individual Participant Data (IPD) Sharing Statement:
    Yes
    Plan to Share IPD:
    Yes
    Studies a U.S. FDA-regulated Drug Product:
    Yes
    Studies a U.S. FDA-regulated Device Product:
    No
    Keywords provided by Pfizer
    Additional relevant MeSH terms:

    Study Results

    Participant Flow

    Recruitment Details This phase 3, randomized, open label study was conducted at 104 sites in 23 countries. A total of 296 participants were randomized, 149 to the lorlatinib arm and 147 to the crizotinib arm.
    Pre-assignment Detail Previously untreated Stage IIIB/IV participants with ALK-positive non-small cell lung cancer were randomized in this study.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Period Title: Treatment Phase
    STARTED 149 147
    COMPLETED 26 83
    NOT COMPLETED 123 64
    Period Title: Treatment Phase
    STARTED 149 147
    COMPLETED 0 0
    NOT COMPLETED 149 147

    Baseline Characteristics

    Arm/Group Title Lorlatinib Crizotinib Total
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Total of all reporting groups
    Overall Participants 149 147 296
    Age (Years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [Years]
    59.1
    (13.12)
    55.6
    (13.52)
    57.4
    (13.41)
    Sex: Female, Male (Count of Participants)
    Female
    84
    56.4%
    91
    61.9%
    175
    59.1%
    Male
    65
    43.6%
    56
    38.1%
    121
    40.9%
    Race/Ethnicity, Customized (Count of Participants)
    White
    72
    48.3%
    72
    49%
    144
    48.6%
    Black or African American
    0
    0%
    1
    0.7%
    1
    0.3%
    Asian
    65
    43.6%
    65
    44.2%
    130
    43.9%
    Missing
    12
    8.1%
    9
    6.1%
    21
    7.1%

    Outcome Measures

    1. Primary Outcome
    Title Progression-Free Survival (PFS) Based on Blinded Independent Central Review (BICR) Assessment
    Description PFS was defined as the time from randomization to the date of the first documentation of progressive disease as assessed by the independent radiologist or death due to any cause, whichever occurred first. PFS (in months) was calculated as (date of event or censoring-randomization+1)/30.4375. Progressive disease is defined per RECIST version 1.1, as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The full analysis (FA) population included all participants who were randomized, and participants were classified according to the treatment assigned at randomization.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 147
    Median (95% Confidence Interval) [Months]
    NA
    9.3
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments The study was designed to test the null hypothesis H0: λ ≥1 versus the alternative hypothesis HA: λ <1, where λ is the hazard ratio (HR; Lorlatinib/Crizotinib). Evaluation of 177 PFS events was required to have at least 90% power to detect a HR of 0.611 using a one-sided stratified log-rank test at a significance level of 0.025 (one-sided), and a 2-look group-sequential design with a Lan-DeMets (O'Brien-Fleming) α-spending function to determine the efficacy boundaries.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments The study was to be considered positive if the 1-sided log-rank test for PFS, stratified for baseline stratification factors (ethnicity and brain metastases) was significant at the 0.0081 level at the cutoff date of this report.
    Method one-sided stratified log-rank
    Comments Stratified by the presence of brain metastases (Yes/No) and ethnic origin (Asian/Non-Asian) at randomization.
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.28
    Confidence Interval (2-Sided) 95%
    0.191 to 0.413
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on Cox Proportional Hazards model stratified by the presence of brain metastases and ethnic origin at randomization. Assuming proportional hazards, a hazard ratio less than 1 indicates a reduction in hazard rate in favor of Lorlatinib.
    2. Secondary Outcome
    Title Overall Survival (OS)
    Description OS was defined as the time from randomization to the date of death due to any cause. OS (in months) was calculated as (date of death or censoring - start date +1)/30.4375. Participants last known to be alive were censored at date of last contact.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The FA population included all participants who were randomized, and participants were classified according to the treatment assigned at randomization.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 147
    Median (95% Confidence Interval) [Months]
    NA
    NA
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.72
    Confidence Interval (2-Sided) 95%
    0.414 to 1.249
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on Cox Proportional Hazards model stratified by the presence of brain metastases and ethnic origin at randomization. Assuming proportional hazards, a hazard ratio less than 1 indicates a reduction in hazard rate in favor of Lorlatinib.
    3. Secondary Outcome
    Title Progression-Free Survival (PFS) Based on Investigator's Assessment
    Description PFS was defined as the time from randomization to the date of the first documentation of progressive disease as assessed by investigator or death due to any cause, whichever occurred first. PFS (in months) was calculated as (date of event or censoring-randomization+1)/30.4375. Progressive disease is defined per RECIST version 1.1, as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The FA population included all participants who were randomized, and participants were classified according to the treatment assigned at randomization.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 147
    Median (95% Confidence Interval) [Months]
    NA
    9.1
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method one-sided stratified log-rank
    Comments Stratified by the presence of brain metastases (Yes/No) and ethnic origin (Asian/Non-Asian) at randomization.
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.21
    Confidence Interval (2-Sided) 95%
    0.144 to 0.307
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on Cox Proportional Hazards model stratified by the presence of brain metastases and ethnic origin at randomization. Assuming proportional hazards, a hazard ratio less than 1 indicates a reduction in hazard rate in favor of Lorlatinib.
    4. Secondary Outcome
    Title Objective Response Rate (ORR) - Percentage of Participants With Objective Response (OR) Based on BICR Assessment
    Description ORR was the percentage of participants with objective response of complete response (CR) or partial response (PR) according to RECIST version 1.1 recorded from randomization until disease progression or start of new anti-cancer therapy. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The FA population included all participants who were randomized, and participants were classified according to the treatment assigned at randomization.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 147
    Number (95% Confidence Interval) [Percentage of participants]
    75.8
    50.9%
    57.8
    39.3%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0005
    Comments
    Method Cochran-Mantel-Haenszel
    Comments Stratified by the presence of brain metastases (Yes/No) and ethnic origin (Asian/Non-Asian) at randomization.
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 2.254
    Confidence Interval (2-Sided) 95%
    1.353 to 3.891
    Parameter Dispersion Type:
    Value:
    Estimation Comments Odds ratio was estimated using Mantel-Haenszel method. An odds ratio larger than 1 indicates better outcome for Lorlatinib relative to Crizotinib. Exact CI was calculated.
    5. Secondary Outcome
    Title Objective Response Rate (ORR) - Percentage of Participants With Objective Response (OR) Based on Investigator's Assessment
    Description ORR was the percentage of participants with objective response of complete response (CR) or partial response (PR) according to RECIST version 1.1 recorded from randomization until disease progression or start of new anti-cancer therapy. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The FA population included all participants who were randomized, and participants were classified according to the treatment assigned at randomization.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 147
    Number (95% Confidence Interval) [Percentage of participants]
    80.5
    54%
    61.9
    42.1%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0002
    Comments
    Method Cochran-Mantel-Haenszel
    Comments Stratified by the presence of brain metastases (Yes/No) and ethnic origin (Asian/Non-Asian) at randomization.
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 2.499
    Confidence Interval (2-Sided) 95%
    1.484 to 4.594
    Parameter Dispersion Type:
    Value:
    Estimation Comments Odds ratio was estimated using Mantel-Haenszel method. An odds ratio larger than 1 indicates better outcome for Lorlatinib relative to Crizotinib. Exact CI was calculated.
    6. Secondary Outcome
    Title Intracranial Objective Response Rate (IC-ORR) - Percentage of Participants With Intracranial Objective Response (IC-OR) Based on BICR Assessment
    Description IC-ORR was the percentage of participant with intracranial objective response of complete response (CR) or partial response (PR) based on intracranial disease in the subset of participants with at least 1 intracranial lesion per RECIST version 1.1 (modified) recorded from randomization until disease progression or start of new anti-cancer therapy. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The subset of the FA population with at least 1 baseline intracranial lesion (based on BICR intracranial assessment).
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 38 40
    Number (95% Confidence Interval) [Percentage of participants]
    65.8
    44.2%
    20.0
    13.6%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method Cochran-Mantel-Haenszel
    Comments Stratified by the presence of brain metastases (Yes/No) and ethnic origin (Asian/Non-Asian) at randomization.
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 8.407
    Confidence Interval (2-Sided) 95%
    2.586 to 27.233
    Parameter Dispersion Type:
    Value:
    Estimation Comments Odds ratio was estimated using Mantel-Haenszel method. An odds ratio larger than 1 indicates better outcome for Lorlatinib relative to Crizotinib. Exact CI was calculated.
    7. Secondary Outcome
    Title Intracranial Time to Progression (IC-TTP) Based on BICR Assessment
    Description IC-TTP based on BICR assessment was defined as the time from date of randomization to the date of the first documentation of progression of intracranial disease, based on either new brain metastases or progression of existing brain metastases.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The FA population included all participants who were randomized, and participants were classified according to the treatment assigned at randomization.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 147
    Median (95% Confidence Interval) [Months]
    NA
    16.6
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method one-sided stratified log-rank
    Comments Stratified by the presence of brain metastases (Yes/No) and ethnic origin (Asian/Non-Asian) at randomization.
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.07
    Confidence Interval (2-Sided) 95%
    0.026 to 0.170
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on Cox Proportional Hazards model stratified by the presence of brain metastases and ethnic origin at randomization. Assuming proportional hazards, a hazard ratio less than 1 indicates a reduction in hazard rate in favor of Lorlatinib.
    8. Secondary Outcome
    Title Duration of Response (DR) Based on BICR Assessment
    Description DR was defined, for participants with a confirmed objective response (OR) of complete response (CR) or partial response (PR) per RECIST version 1.1, as the time from the first documentation of OR to the first documentation of progressive disease (PD) or death due to any cause, whichever occurred first. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions. PD was defined as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants with a confirmed objective response (complete response or partial response) per RECIST version 1.1 in the FA population.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 113 85
    Median (95% Confidence Interval) [Months]
    NA
    11.0
    9. Secondary Outcome
    Title Intracranial Duration of Response (IC-DR) Based on BICR Assessment
    Description IC-DR was defined, for participants with a confirmed intracranial objective response (OR) of complete response (CR) or partial response (PR) per RECIST version 1.1, as the time from the first documentation of intracranial OR to the first documentation of intracranial progressive disease (PD) or death due to any cause, whichever occurred first. CR was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions. PD was defined as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of the longest dimensions of the target lesions taking as a reference the smallest sum of the longest dimensions recorded since the treatment started, or the appearance of 1 or more new lesions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants with brain metastases at baseline and confirmed intracranial complete response or partial response in the FA population.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 25 8
    Median (95% Confidence Interval) [Months]
    NA
    9.4
    10. Secondary Outcome
    Title Time to Tumor Response (TTR) Based on BICR Assessment
    Description TTR based on BICR assessment was defined, for participants with a confirmed objective response, as the time from the date of randomization to the first documentation of objective response (complete response or partial response) which was subsequently confirmed. Complete response was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). Partial response was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants with confirmed complete response or partial response in the FA population.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 113 85
    Median (Inter-Quartile Range) [Months]
    1.8
    1.8
    11. Secondary Outcome
    Title Intracranial Time to Tumor Response (IC-TTR) Based on BICR Assessment
    Description IC-TTR was defined, for participants with a confirmed intracranial objective response, as the time from the date of randomization to the first documentation of intracranial objective response (complete response or partial response) which was subsequently confirmed. Complete response was defined as complete disappearance of all target lesions and non-target disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). Partial response was defined as at least a 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants with brain metastases at baseline and confirmed intracranial complete response or partial response in the FA population.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 25 8
    Median (Inter-Quartile Range) [Months]
    1.9
    1.8
    12. Secondary Outcome
    Title PFS2 Based on Investigator's Assessment
    Description PFS2 was defined as the time from randomization to the date of progression of disease on first subsequent systemic anti-cancer therapy, or death from any cause, whichever occurred first
    Time Frame From time of Study Start up to 45 months

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    13. Secondary Outcome
    Title Number of Participants With Treatment-Emergent Adverse Events (AEs; All-Causality and Treatment-Related)
    Description An AE was an untoward medical occurrence in a participant who received study treatment without regard to possibility of causal relationship. Serious adverse event (SAE) was an AE resulting in any of the following outcomes: death, life-threatening experience, initial or prolonged inpatient hospitalization, persistent or significant disability/incapacity, congenital anomaly/birth defect. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. All AEs in the table below were treatment-emergent AEs. Grade 3 and 4 AEs in the table below indicated severe AE and life-threatening consequences respectively; Grade 5 indicated death due to AE. Treatment-related AEs were determined by investigators.
    Time Frame From time of Study Start up to 33 months

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population included all participants who received at least 1 dose of study drug. Participants were classified according to the treatment assigned at randomization unless the incorrect treatment(s) were received throughout the dosing period, in which case participants were classified according to the first study treatment received.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 142
    AEs (all-causality)
    149
    100%
    140
    95.2%
    AEs (treatment-related)
    144
    96.6%
    133
    90.5%
    SAEs (all-causality)
    51
    34.2%
    39
    26.5%
    SAEs (treatment-related)
    12
    8.1%
    7
    4.8%
    Maximum Grade 3 or 4 AEs (all-causality)
    108
    72.5%
    79
    53.7%
    Maximum Grade 3 or 4 AEs (treatment-related)
    83
    55.7%
    52
    35.4%
    Maximum Grade 5 AEs (all-causality)
    7
    4.7%
    7
    4.8%
    Maximum Grade 5 AEs (treatment-related)
    2
    1.3%
    0
    0%
    AEs causing study discontinuation (all-causality)
    7
    4.7%
    8
    5.4%
    AEs causing study discontinuation (treatment-related)
    2
    1.3%
    0
    0%
    AEs causing study treatment discontinuation (all-causality)
    10
    6.7%
    13
    8.8%
    AEs causing study treatment discontinuation (treatment-related)
    7
    4.7%
    7
    4.8%
    AEs causing dose reduction or temporary discontinuation (all-causality)
    79
    53%
    71
    48.3%
    AEs causing dose reduction or temporary discontinuation (treatment-related)
    60
    40.3%
    54
    36.7%
    14. Secondary Outcome
    Title Number of Participants With Changes in Hematology Laboratory Parameters From Baseline Maximum NCI-CTCAE Grade Lower Than 3 to Postbaseline Maximum Grade 3 or Grade 4
    Description Laboratory test results were graded according to National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.03. The CTCAE displays Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grades 1 and 2 indicate mild and moderate AE, respectively; Grades 3 and 4 indicate severe AE and life-threatening consequences respectively; Grade 5 indicates death due to AE. Participants with laboratory test abnormalities were summarized according to the worst grade for each laboratory test result. All participants meeting the postbaseline grade criteria in the table below had baseline maximum grades lower than 3.
    Time Frame From Baseline up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants in the safety analysis population who had at least one on-study assessment for the parameter of interest.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 141
    Anemia (Postbaseline Maximum Grade 3)
    3
    2%
    4
    2.7%
    Anemia (Postbaseline Maximum Grade 4)
    NA
    NaN
    NA
    NaN
    Hemoglobin increased (Postbaseline Maximum Grade 3)
    0
    0%
    0
    0%
    Hemoglobin increased (Postbaseline Maximum Grade 4)
    NA
    NaN
    NA
    NaN
    Lymphocyte count decreased (Postbaseline Maximum Grade 3)
    2
    1.3%
    7
    4.8%
    Lymphocyte count decreased (Postbaseline Maximum Grade 4)
    2
    1.3%
    1
    0.7%
    Lymphocyte count increased (Postbaseline Maximum Grade 3)
    0
    0%
    0
    0%
    Lymphocyte count increased (Postbaseline Maximum Grade 4)
    NA
    NaN
    NA
    NaN
    Neutrophil count decreased (Postbaseline Maximum Grade 3)
    1
    0.7%
    19
    12.9%
    Neutrophil count decreased (Postbaseline Maximum Grade 4)
    1
    0.7%
    4
    2.7%
    Platelet count decreased (Postbaseline Maximum Grade 3)
    0
    0%
    1
    0.7%
    Platelet count decreased (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    White blood cell decreased (Postbaseline Maximum Grade 3)
    0
    0%
    5
    3.4%
    White blood cell decreased (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    15. Secondary Outcome
    Title Number of Participants With Changes in Chemistry Laboratory Parameters From Baseline Maximum NCI-CTCAE Grade Lower Than 3 to Postbaseline Maximum Grade 3 or Grade 4
    Description Laboratory test results were graded according to National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.03. The CTCAE displays Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grades 1 and 2 indicate mild and moderate AE, respectively; Grades 3 and 4 indicate severe AE and life-threatening consequences respectively; Grade 5 indicates death due to AE. Participants with laboratory test abnormalities were summarized according to the worst grade for each laboratory test result. All participants meeting the postbaseline grade criteria in the table below had baseline maximum grades lower than 3.
    Time Frame From Baseline up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants in the safety analysis population who had at least one on-study assessment for the parameter of interest.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 141
    Alanine aminotransferase increased (Postbaseline Maximum Grade 3)
    4
    2.7%
    5
    3.4%
    Alanine aminotransferase increased (Postbaseline Maximum Grade 4)
    0
    0%
    1
    0.7%
    Alkaline phosphate increased (Postbaseline Maximum Grade 3)
    0
    0%
    1
    0.7%
    Alkaline phosphate increased (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Aspartate aminotransferase increased (Postbaseline Maximum Grade 3)
    3
    2%
    4
    2.7%
    Aspartate aminotransferase increased (Postbaseline Maximum Grade 4)
    0
    0%
    1
    0.7%
    Blood bilirubin increased (Postbaseline Maximum Grade 3)
    0
    0%
    0
    0%
    Blood bilirubin increased (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Creatine kinase increased (Postbaseline Maximum Grade 3)
    3
    2%
    5
    3.4%
    Creatine kinase increased (Postbaseline Maximum Grade 4)
    0
    0%
    2
    1.4%
    Creatinine increased (Postbaseline Maximum Grade 3)
    1
    0.7%
    3
    2%
    Creatinine increased (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Gamma glutamyl transferase increased (Postbaseline Maximum Grade 3)
    9
    6%
    8
    5.4%
    Gamma glutamyl transferase increased (Postbaseline Maximum Grade 4)
    0
    0%
    1
    0.7%
    Hypercalcemia (Postbaseline Maximum Grade 3)
    0
    0%
    0
    0%
    Hypercalcemia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Hyperglycemia (Postbaseline Maximum Grade 3)
    9
    6%
    3
    2%
    Hyperglycemia (Postbaseline Maximum Grade 4)
    1
    0.7%
    0
    0%
    Hyperkalemia (Postbaseline Maximum Grade 3)
    2
    1.3%
    3
    2%
    Hyperkalemia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Hypermagnesemia (Postbaseline Maximum Grade 3)
    2
    1.3%
    1
    0.7%
    Hypermagnesemia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Hypernatremia (Postbaseline Maximum Grade 3)
    0
    0%
    0
    0%
    Hypernatremia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Hypoalbuminemia (Postbaseline Maximum Grade 3)
    1
    0.7%
    9
    6.1%
    Hypoalbuminemia (Postbaseline Maximum Grade 4)
    NA
    NaN
    NA
    NaN
    Hypocalcemia (Postbaseline Maximum Grade 3)
    1
    0.7%
    0
    0%
    Hypocalcemia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Hypoglycemia (Postbaseline Maximum Grade 3)
    0
    0%
    0
    0%
    Hypoglycemia (Postbaseline Maximum Grade 4)
    0
    0%
    1
    0.7%
    Hypokalemia (Postbaseline Maximum Grade 3)
    0
    0%
    3
    2%
    Hypokalemia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Hypomagnesemia (Postbaseline Maximum Grade 3)
    0
    0%
    0
    0%
    Hypomagnesemia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Hyponatremia (Postbaseline Maximum Grade 3)
    5
    3.4%
    10
    6.8%
    Hyponatremia (Postbaseline Maximum Grade 4)
    0
    0%
    1
    0.7%
    Hypophosphatemia (Postbaseline Maximum Grade 3)
    3
    2%
    4
    2.7%
    Hypophosphatemia (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    Lipase increased (Postbaseline Maximum Grade 3)
    8
    5.4%
    6
    4.1%
    Lipase increased (Postbaseline Maximum Grade 4)
    3
    2%
    1
    0.7%
    Serum amylase increased (Postbaseline Maximum Grade 3)
    1
    0.7%
    2
    1.4%
    Serum amylase increased (Postbaseline Maximum Grade 4)
    0
    0%
    0
    0%
    16. Secondary Outcome
    Title Number of Participants With Changes in Lipid Laboratory Parameters From Baseline Maximum NCI-CTCAE Grade Lower Than 3 to Postbaseline Maximum Grade 3 or Grade 4
    Description Laboratory test results were graded according to National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.03. The CTCAE displays Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grades 1 and 2 indicate mild and moderate AE, respectively; Grades 3 and 4 indicate severe AE and life-threatening consequences respectively; Grade 5 indicates death due to AE. Participants with laboratory test abnormalities were summarized according to the worst grade for each laboratory test result. All participants meeting the postbaseline grade criteria in the table below had baseline maximum grades lower than 3.
    Time Frame From Baseline up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants in the safety analysis population who had at least one on-study assessment for the parameter of interest.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 141
    Cholesterol high (Postbaseline Maximum Grade 3)
    26
    17.4%
    0
    0%
    Cholesterol high (Postbaseline Maximum Grade 4)
    3
    2%
    0
    0%
    Hypertriglyceridemia (Postbaseline Maximum Grade 3)
    20
    13.4%
    0
    0%
    Hypertriglyceridemia (Postbaseline Maximum Grade 4)
    13
    8.7%
    0
    0%
    17. Secondary Outcome
    Title Number of Participant With Vital Signs and Body Weight Data Meeting Pre-defined Criteria
    Description Vital signs data included pulse, systolic blood pressure, and diastolic blood pressure. Measurements were only provided once per timepoint. If multiple assessments were provided per timepoint, the maximum value were used for reporting. The pre-defined criteria of vital sign and body weight data were as follows: maximum pulse rate >120 beats per minute (bpm); minimum pulse rate <50 bpm; maximum increase in pulse rate ≥30 bpm; maximum decrease in pulse rate ≥30 bpm; increase in systolic blood Pressure ≥40 mmHg; decrease in systolic blood pressure ≥40 mmHg; decrease in systolic blood pressure ≥60 mmHg; increase in diastolic blood pressure ≥20 mmHg; decrease in diastolic blood pressure ≥20 mmHg; decrease in diastolic blood pressure ≥40 mmHg; increase in body weight ≥10%; increase in body weight ≥20%; decrease in body weight ≥10%.
    Time Frame From Baseline up to 33 months

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the safety analysis population. "Number Analyzed" included all participants in the safety analysis population with evaluable data against the pre-defined criteria.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 142
    Sitting pulse rate: pulse >120 bpm
    7
    4.7%
    1
    0.7%
    Sitting pulse rate: pulse <50 bpm
    4
    2.7%
    22
    15%
    Sitting pulse rate: change ≥30 bpm increase
    24
    16.1%
    3
    2%
    Sitting pulse rate: change ≥30 bpm decrease
    17
    11.4%
    47
    32%
    Sitting systolic blood pressure: change ≥40 mmHg increase
    22
    14.8%
    3
    2%
    Sitting systolic blood pressure: change ≥40 mmHg decrease
    6
    4%
    14
    9.5%
    Sitting systolic blood pressure: change ≥60 mmHg decrease
    0
    0%
    1
    0.7%
    Sitting diastolic blood pressure: change ≥20 mmHg increase
    41
    27.5%
    18
    12.2%
    Sitting diastolic blood pressure: change ≥20 mmHg decrease
    26
    17.4%
    52
    35.4%
    Sitting diastolic blood pressure: change ≥40 mmHg decrease
    0
    0%
    1
    0.7%
    Body weight (kg): percent change from baseline ≥10% increase
    99
    66.4%
    39
    26.5%
    Body weight (kg): percent change from baseline ≥20% increase
    35
    23.5%
    3
    2%
    Body weight (kg): percent change from baseline ≥10% decrease
    6
    4%
    12
    8.2%
    18. Secondary Outcome
    Title Number of Participant With Maximum Increase From Baseline in Electrocardiogram (ECG) Data Meeting Pre-defined Criteria
    Description Baseline was defined as the last assessment performed on or prior to date of the first dose of study treatment. Triplicate ECGs were collected in the study and the average of the replicate assessments were used for summary analysis. The pre-defined criteria of ECG data were as follows: change from baseline in QTcF ≥60 msec, ≥30 msec but <60 msec, <30 msec; change from baseline in QTcB ≥60 msec, ≥30 msec but <60 msec, <30 msec; PR change ≥50% if absolute baseline value was <200 msec; PR change ≥25% if absolute baseline value was ≥200 msec; QRS change ≥50% if absolute baseline value was <100 msec; QRS change ≥25% if absolute baseline value was ≥100 msec.
    Time Frame From Baseline up to 33 months

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the safety analysis population. "Number Analyzed" included all participants in the safety analysis population with evaluable data against the pre-defined criteria.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 142
    Change of QTcF ≥60 msec
    5
    3.4%
    16
    10.9%
    30 msec ≤ Change of QTcF <60 msec
    49
    32.9%
    41
    27.9%
    Change of QTcF <30 msec
    92
    61.7%
    81
    55.1%
    Change of QTcB ≥60 msec
    10
    6.7%
    15
    10.2%
    30 msec ≤ Change of QTcB <60 msec
    49
    32.9%
    26
    17.7%
    Change of QTcB <30 msec
    90
    60.4%
    100
    68%
    PR change ≥50% if absolute baseline value was <200 msec
    8
    5.4%
    4
    2.7%
    PR change ≥25% if absolute baseline value was ≥200 msec
    0
    0%
    0
    0%
    QRS change ≥50% if absolute baseline value was <100 msec
    2
    1.3%
    3
    2%
    QRS change ≥25% if absolute baseline value was ≥100 msec
    2
    1.3%
    0
    0%
    19. Secondary Outcome
    Title Number of Participants With Maximum Decrease From Baseline Greater Than or Equal to 20 Points in Left Ventricular Ejection Fraction (LVEF) Percentage
    Description In this outcome measure, baseline was defined as the last assessment on or prior to the date of the first dose of study treatment. Decrease from baseline was an absolute difference between baseline and observed value.
    Time Frame From Baseline up to 33 months

    Outcome Measure Data

    Analysis Population Description
    All participants in the safety analysis population who had at least one on-study assessment for the parameter of interest.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 143 133
    Count of Participants [Participants]
    2
    1.3%
    1
    0.7%
    20. Secondary Outcome
    Title Change From Baseline in Total Scores of Beck Depression Inventory (BDI)-II (Mood Assessment) Across Time
    Description BDI-II (Mood Assessment) is a 21 item self-reported scale, with each item rated by participants on a 4-point scale (ranging from 0-3). The scale includes items capturing mood, (loss of pleasure, sadness, and irritability), suicidal ideation, and cognitive signs (punitive thoughts, self-criticism, self-dislike pessimism, poor concentration) as well as somatic signs (appetite, sleep, fatigue, libido). Scores were obtained by adding up the total points from the series of answers. The total score ranged from 0 to 63, with higher total scores indicating more severe depressive symptoms. The standardized cutoffs are as follows: 0-13: minimal depression; 14-19: mild depression; 20-28: moderate depression; 29-63: severe depression.
    Time Frame Baseline, Cycle 2 Day 1 (C2D1), C3D1, C4D1, C5D1, C6D1, C8D1, C10D1, C12D1, C14D1, C16D1, C18D1, C20D1, C22D1, C24D1, C26D1, C28D1, C30D1, C32D1, C34D1, C36D1, C38D1 and End of Treatment

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the safety analysis population within each treatment group. "Number Analyzed" included all participants with at least a baseline and postbaseline assessment at the visit.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 147
    Cycle 2 Day 1
    -1.8
    (5.52)
    -1.2
    (5.62)
    Cycle 3 Day 1
    -2.5
    (5.85)
    -2.7
    (5.78)
    Cycle 4 Day 1
    -2.9
    (5.73)
    -3.6
    (6.60)
    Cycle 5 Day 1
    -2.5
    (6.28)
    -3.0
    (6.24)
    Cycle 6 Day 1
    -2.6
    (6.61)
    -2.9
    (6.65)
    Cycle 8 Day 1
    -3.2
    (6.18)
    -1.9
    (6.56)
    Cycle 10 Day 1
    -3.0
    (6.30)
    -2.6
    (5.98)
    Cycle 12 Day 1
    -3.6
    (6.05)
    -2.1
    (6.23)
    Cycle 14 Day 1
    -3.3
    (6.82)
    -2.5
    (5.57)
    Cycle 16 Day 1
    -3.3
    (6.87)
    -2.3
    (5.77)
    Cycle 18 Day 1
    -3.7
    (7.26)
    -2.6
    (4.30)
    Cycle 20 Day 1
    -3.3
    (6.24)
    -2.4
    (4.62)
    Cycle 22 Day 1
    -3.2
    (6.95)
    -2.5
    (4.82)
    Cycle 24 Day 1
    -3.1
    (5.97)
    2.3
    (9.31)
    Cycle 26 Day 1
    -3.2
    (6.87)
    -0.8
    (3.81)
    Cycle 28 Day 1
    -3.0
    (6.53)
    0.2
    (7.65)
    Cycle 30 Day 1
    -4.8
    (8.00)
    1.3
    (3.20)
    Cycle 32 Day 1
    -5.0
    (14.14)
    7.5
    (9.19)
    Cycle 34 Day 1
    -2.3
    (12.42)
    0.0
    Cycle 36 Day 1
    0.0
    End of Treatment
    0.8
    (4.49)
    -0.2
    (7.45)
    21. Secondary Outcome
    Title Number of Participants With Suicidal Ideation and Suicidal Behavior Across Time
    Description Suicidal ideation and behaviors were assessed by the Columbia Suicide Severity Rating Scale (C-SSRS). The C-SSRS is a unique, simple and short method of assessing both behavior and ideation that tracks all suicidal events and provides a summary of suicidality. It assesses the lethality of attempts and other features of ideation (frequency, duration, controllability, reasons for ideation, and deterrents), all of which are significantly predictive of completed suicide.
    Time Frame Baseline, Cycle 2 Day 1 (C2D1), C3D1, C4D1, C5D1, C6D1, C8D1, C10D1, C12D1, C14D1, C16D1, C18D1, C20D1, C22D1, C24D1, C26D1, C28D1, C30D1, C32D1, C34D1, C36D1, C38D1 and End of Treatment

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the safety analysis population within each treatment group. "Number Analyzed" included all participants with assessment scores at each specified time point.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 149 142
    Baseline
    3
    2%
    7
    4.8%
    Cycle 2 Day 1
    0
    0%
    5
    3.4%
    Cycle 3 Day 1
    1
    0.7%
    1
    0.7%
    Cycle 4 Day 1
    0
    0%
    1
    0.7%
    Cycle 5 Day 1
    0
    0%
    1
    0.7%
    Cycle 6 Day 1
    0
    0%
    2
    1.4%
    Cycle 8 Day 1
    0
    0%
    2
    1.4%
    Cycle 10 Day 1
    0
    0%
    1
    0.7%
    Cycle 12 Day 1
    0
    0%
    0
    0%
    Cycle 14 Day 1
    0
    0%
    0
    0%
    Cycle 16 Day 1
    0
    0%
    0
    0%
    Cycle 18 Day 1
    0
    0%
    0
    0%
    Cycle 20 Day 1
    0
    0%
    0
    0%
    Cycle 22 Day 1
    0
    0%
    0
    0%
    Cycle 24 Day 1
    0
    0%
    1
    0.7%
    Cycle 26 Day 1
    1
    0.7%
    0
    0%
    Cycle 28 Day 1
    0
    0%
    0
    0%
    Cycle 30 Day 1
    0
    0%
    0
    0%
    Cycle 32 Day 1
    0
    0%
    0
    0%
    Cycle 34 Day 1
    0
    0%
    0
    0%
    Cycle 36 Day 1
    0
    0%
    0
    0%
    Cycle 38 Day 1
    0
    0%
    End of Treatment
    0
    0%
    0
    0%
    22. Secondary Outcome
    Title Change From Baseline in Global Quality of Life (QOL), Functional Scales and Symptoms Scales as Assessed by the European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC-QLQ-C30) During Overall Treatment
    Description The EORTC QLQ C30 consists of 30 questions and includes 5 functional scales (physical, role, cognitive, emotional, and social); a global health status/global quality of life scale; 3 symptom scales (fatigue, pain, nausea and vomiting); and 6 single items that assess additional symptoms (dyspnea, appetite loss, sleep disturbance, constipation, and diarrhea) and financial impact. All scales and single item measures range in score from 0 to 100. Higher scores on the functional scales represent higher levels of functioning. Higher scores on the global health status/quality of life scale represent higher health status/quality of life. Higher scores on symptom scales/items represent a greater presence of symptoms.
    Time Frame From Baseline up to Cycle 38 Day 1

    Outcome Measure Data

    Analysis Population Description
    Participants from the FA population who completed a baseline (last patient-reported outcome (PRO) assessment prior to the first dose of study treatment) and at least 1 post-baseline PRO assessment.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 148 140
    Global QOL
    8.60
    3.95
    Physical Functioning
    4.84
    2.82
    Role Functioning
    6.86
    4.78
    Emotional Functioning
    8.77
    6.20
    Cognitive Functioning
    -4.20
    -1.02
    Social Functioning
    7.00
    4.72
    Fatigue
    -9.93
    -4.26
    Nausea and Vomiting
    -4.35
    3.51
    Pain
    -4.60
    -5.76
    Dyspnoea
    -7.02
    -8.75
    Insomnia
    -17.34
    -9.39
    Appetite Loss
    -13.15
    -3.95
    Constipation
    -2.40
    2.53
    Diarrhea
    -0.92
    11.12
    Financial Difficulties
    -6.79
    -5.74
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Global QOL. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0096
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 4.65
    Confidence Interval (2-Sided) 95%
    1.14 to 8.16
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Physical Functioning. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.1897
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 2.02
    Confidence Interval (2-Sided) 95%
    -1.01 to 5.05
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 3
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Role Functioning. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.2999
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 2.09
    Confidence Interval (2-Sided) 95%
    -1.87 to 6.04
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 4
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Emotional Functioning. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0553
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 2.58
    Confidence Interval (2-Sided) 95%
    -0.06 to 5.22
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 5
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Cognitive Functioning. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0588
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -3.18
    Confidence Interval (2-Sided) 95%
    -6.47 to 0.12
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 6
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Social Functioning. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.2118
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 2.27
    Confidence Interval (2-Sided) 95%
    -1.30 to 5.85
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 7
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Fatigue. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0032
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -5.67
    Confidence Interval (2-Sided) 95%
    -9.42 to -1.92
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 8
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Nausea and Vomiting. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -7.86
    Confidence Interval (2-Sided) 95%
    -9.86 to -5.86
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 9
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Pain. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.5310
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 1.16
    Confidence Interval (2-Sided) 95%
    -2.49 to 4.82
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 10
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Dyspnoea. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.3602
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 1.72
    Confidence Interval (2-Sided) 95%
    -1.98 to 5.43
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 11
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Insomnia. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -7.95
    Confidence Interval (2-Sided) 95%
    -11.25 to -4.64
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 12
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Appetite Loss. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -9.21
    Confidence Interval (2-Sided) 95%
    -11.80 to -6.62
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 13
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Constipation. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0198
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -4.93
    Confidence Interval (2-Sided) 95%
    -9.07 to -0.79
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 14
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Diarrhea. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -12.03
    Confidence Interval (2-Sided) 95%
    -15.49 to -8.58
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 15
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-C30 Financial Difficulties. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.5947
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -1.04
    Confidence Interval (2-Sided) 95%
    -4.90 to 2.82
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    23. Secondary Outcome
    Title Change From Baseline in Lung Cancer Symptoms as Assessed by the EORTC Quality of Life Questionnaire-Lung Cancer 13 (QLQ- LC13) During Overall Treatment
    Description The EORTC QLQ LC13 consists of 13 questions and includes 1 multi-item scale and 9 single items assessing symptoms (dyspnea, cough, haemoptysis, and site-specific pain), side effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication use. The scale scores rang from 0 to 100, with higher scores indicating higher ("worse") level of symptoms.
    Time Frame From Baseline up to Cycle 38 Day 1

    Outcome Measure Data

    Analysis Population Description
    Participants from the FA population who completed a baseline (last patient-reported outcome (PRO) assessment prior to the first dose of study treatment) and at least 1 post-baseline PRO assessment.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 146 139
    Dyspnoea
    -4.36
    -4.90
    Coughing
    -21.21
    -16.66
    Haemoptysis
    -2.53
    -2.65
    Sore Mouth
    0.56
    -0.60
    Dysphagia
    -1.35
    0.16
    Peripheral Neuropathy
    11.56
    6.20
    Alopecia
    1.61
    1.81
    Pain in Chest
    -9.54
    -9.01
    Pain in Arm or Shoulder
    -6.93
    -7.38
    Pain in Other Parts
    -2.31
    -5.67
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Dyspnoea. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.7254
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 0.55
    Confidence Interval (2-Sided) 95%
    -2.51 to 3.60
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Coughing. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0115
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -4.55
    Confidence Interval (2-Sided) 95%
    -8.06 to -1.03
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 3
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Haemoptysis. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.7824
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 0.12
    Confidence Interval (2-Sided) 95%
    -0.75 to 0.99
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 4
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Sore Mouth. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.2988
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 1.16
    Confidence Interval (2-Sided) 95%
    -1.04 to 3.36
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 5
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Dysphagia. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.1916
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -1.51
    Confidence Interval (2-Sided) 95%
    -3.79 to 0.76
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 6
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Peripheral Neuropathy. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0279
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 5.37
    Confidence Interval (2-Sided) 95%
    0.59 to 10.15
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 7
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Alopecia. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.9162
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -0.20
    Confidence Interval (2-Sided) 95%
    -3.89 to 3.49
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 8
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Pain in Chest. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.6698
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value -0.53
    Confidence Interval (2-Sided) 95%
    -2.96 to 1.90
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 9
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Pain in Arm or Shoulder. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.8035
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 0.45
    Confidence Interval (2-Sided) 95%
    -3.13 to 4.04
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 10
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments This is the analysis for QLQ-LC13 Pain in Other Parts. Estimated change from baseline was based on random intercept random slope mixed-effects model with an intercept term, treatment, time (as a continuous variable), treatment-by-time, baseline and randomization stratification factors as covariates.
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.1143
    Comments
    Method Mixed Models Analysis
    Comments
    Method of Estimation Estimation Parameter Mean Difference (Net)
    Estimated Value 3.36
    Confidence Interval (2-Sided) 95%
    -0.82 to 7.55
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    24. Secondary Outcome
    Title Change From Baseline in Health Status as Assessed by EuroQol 5 Dimension 5 Level (EQ-5D-5L) - Visual Analogue Scale (VAS) Across Time
    Description The EuroQol EQ-5D-5L is a participant-completed questionnaire designed to assess health status. There are 2 components to the EuroQol EQ-5D-5L: a descriptive system in which individuals rate their level of problems (none, slight, moderate, severe, extreme/unable) in 5 areas (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) and a VAS. The VAS component rates current health state on a scale from 0 (worst imaginable health state) to 100 (best imaginable health state) and higher scores indicate better health state.
    Time Frame Baseline, Day 1 of all cycles from Cycle 2 to Cycle 38, and End of Treatment

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the EQ-5D-5L PRO analysis population within each treatment group. "Number Analyzed" included all participants with at least a baseline and postbaseline assessment at the visit.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 147 138
    Cycle 2 Day 1
    6.0
    3.1
    Cycle 3 Day 1
    6.0
    5.3
    Cycle 4 Day 1
    7.1
    5.6
    Cycle 5 Day 1
    4.3
    5.1
    Cycle 6 Day 1
    6.5
    4.5
    Cycle 7 Day 1
    5.5
    4.0
    Cycle 8 Day 1
    6.7
    4.2
    Cycle 9 Day 1
    6.4
    3.9
    Cycle 10 Day 1
    7.2
    3.9
    Cycle 11 Day 1
    8.1
    2.0
    Cycle 12 Day 1
    7.6
    4.8
    Cycle 13 Day 1
    7.5
    5.8
    Cycle 14 Day 1
    6.7
    2.3
    Cycle 15 Day 1
    6.1
    1.5
    Cycle 16 Day 1
    6.9
    2.4
    Cycle 17 Day 1
    8.3
    1.1
    Cycle 18 Day 1
    8.7
    1.3
    Cycle 19 Day 1
    6.8
    2.8
    Cycle 20 Day 1
    7.9
    3.4
    Cycle 21 Day 1
    7.2
    3.3
    Cycle 22 Day 1
    4.5
    1.9
    Cycle 23 Day 1
    5.2
    -0.6
    Cycle 24 Day 1
    4.9
    -3.0
    Cycle 25 Day 1
    4.0
    1.4
    Cycle 26 Day 1
    7.9
    2.0
    Cycle 27 Day 1
    4.6
    3.0
    Cycle 28 Day 1
    6.0
    2.7
    Cycle 29Day 1
    7.4
    8.2
    Cycle 30 Day 1
    9.1
    2.8
    Cycle 31 Day 1
    7.8
    0.0
    Cycle 32 Day 1
    7.9
    2.5
    Cycle 33 Day 1
    4.4
    -2.5
    Cycle 34 Day 1
    -0.5
    20.0
    Cycle 35 Day 1
    0.0
    20.0
    Cycle 36 Day 1
    -4.5
    15.0
    Cycle 37 Day 1
    -17.5
    Cycle 38 Day 1
    25.0
    End of Treatment
    -3.7
    -1.3
    25. Secondary Outcome
    Title Change From Baseline in Health Status as Assessed by EuroQol 5 Dimension 5 Level (EQ-5D-5L) - Index Across Time
    Description The EuroQol EQ-5D-5L is a participant-completed questionnaire designed to assess health status. There are 2 components to the EuroQol EQ-5D-5L: a descriptive system in which individuals rate their level of problems (none, slight, moderate, severe, extreme/unable) in 5 areas (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) and a VAS in which participants rate their overall health status from 0 (worst imaginable) to 100 (best imaginable). EQ-5D summary index is obtained with a formula that weights each level of the 5 dimensions. The index-based score is interpreted along a continuum of 0 (death) to 1 (perfect health).
    Time Frame Baseline, Day 1 of all cycles from Cycle 2 to Cycle 38, and End of Treatment

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the PRO analysis population within each treatment group. "Number Analyzed" included all participants with at least a baseline and postbaseline assessment at the visit.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 148 140
    Cycle 2 Day 1
    0.078
    0.064
    Cycle 3 Day 1
    0.071
    0.101
    Cycle 4 Day 1
    0.089
    0.098
    Cycle 5 Day 1
    0.047
    0.100
    Cycle 6 Day 1
    0.077
    0.070
    Cycle 7 Day 1
    0.062
    0.046
    Cycle 8 Day 1
    0.074
    0.061
    Cycle 9 Day 1
    0.060
    0.050
    Cycle 10 Day 1
    0.074
    0.050
    Cycle 11 Day 1
    0.083
    0.057
    Cycle 12 Day 1
    0.080
    0.049
    Cycle 13 Day 1
    0.081
    0.055
    Cycle 14 Day 1
    0.091
    0.021
    Cycle 15 Day 1
    0.077
    0.036
    Cycle 16 Day 1
    0.091
    0.051
    Cycle 17 Day 1
    0.098
    -0.016
    Cycle 18 Day 1
    0.103
    0.021
    Cycle 19 Day 1
    0.100
    0.027
    Cycle 20 Day 1
    0.103
    0.025
    Cycle 21 Day 1
    0.068
    0.040
    Cycle 22 Day 1
    0.079
    0.052
    Cycle 23 Day 1
    0.060
    0.006
    Cycle 24 Day 1
    0.082
    -0.053
    Cycle 25 Day 1
    0.047
    0.045
    Cycle 26 Day 1
    0.065
    0.039
    Cycle 27 Day 1
    0.042
    0.021
    Cycle 28 Day 1
    0.024
    -0.040
    Cycle 29 Day 1
    0.046
    0.041
    Cycle 30 Day 1
    0.099
    -0.028
    Cycle 31 Day 1
    0.086
    -0.024
    Cycle 32 Day 1
    0.058
    -0.139
    Cycle 33 Day 1
    0.058
    -0.024
    Cycle 34 Day 1
    0.062
    0.042
    Cycle 35 Day 1
    0.129
    0.042
    Cycle 36 Day 1
    -0.027
    0.042
    Cycle 37 Day 1
    0.056
    Cycle 38 Day 1
    0.232
    End of Treatment
    -0.033
    0.001
    26. Secondary Outcome
    Title Time to Deterioration (TTD) in Participant Reported Pain in Chest, Dyspnea, or Cough From QLQ-LC13
    Description The EORTC QLQ LC13 consists of 13 questions and includes 1 multi-item scale and 9 single items assessing symptoms (dyspnea, cough, haemoptysis, and site-specific pain), side effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication use. The scale scores rang from 0 to 100, with higher scores indicating higher ("worse") level of symptoms. TTD in pain in chest, dyspnea, or cough was defined as the time from randomization to the first time the participant's score showed a 10 point or greater increase after baseline in any of the 3 symptoms. TTD in months was calculated as (date of deterioration or censoring - randomization date +1)/30.4375.
    Time Frame From Baseline up to 33 months

    Outcome Measure Data

    Analysis Population Description
    Participants in the EORTC QLQ-LC13 PRO analysis population with change from baseline scores within each treatment group and subscale.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 146 139
    Median (95% Confidence Interval) [Months]
    3.3
    3.7
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Lorlatinib, Crizotinib
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.7293
    Comments
    Method one-sided stratified log-rank
    Comments Stratified by the presence of brain metastases (Yes/No) and ethnic origin (Asian/Non-Asian) at randomization.
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 1.09
    Confidence Interval (2-Sided) 95%
    0.822 to 1.444
    Parameter Dispersion Type:
    Value:
    Estimation Comments Based on Cox Proportional Hazards model stratified by the presence of brain metastases and ethnic origin at randomization. Assuming proportional hazards, a hazard ratio less than 1 indicates a reduction in hazard rate in favor of Lorlatinib.
    27. Secondary Outcome
    Title Number of Participant With ALK Domain Mutation in Plasma Circulating Nucleic Acid (CNA) Analysis at Screening, Cycle 2 Day 1 and Cycle 7 Day 1
    Description The analysis of anaplastic lymphoma kinase (ALK) domain mutation in plasma CNA was performed by next-generation sequencing (NGS) and the number of participants with one or more ALK mutations at Screening, Cycle 2 Day 1 and Cycle 7 Day 1 is presented here.
    Time Frame at Screening, Cycle 2 Day 1 and Cycle 7 Day 1

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the plasma CNA analysis population within each treatment group at screening. "Number Analyzed" included all participants in the plasma CNA analysis population within each treatment group at the specified visit.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 130 125
    ≥1 ALK Mutation Detected
    5
    3.4%
    6
    4.1%
    No ALK Mutation Detected
    88
    59.1%
    91
    61.9%
    No Circulating Free Deoxyribonucleic Acid (cfDNA) Detected
    32
    21.5%
    25
    17%
    Other (Sample failed analysis, uninformative, or not analyzed.)
    5
    3.4%
    3
    2%
    ≥1 ALK Mutation Detected
    2
    1.3%
    3
    2%
    No ALK Mutation Detected
    66
    44.3%
    55
    37.4%
    No Circulating Free Deoxyribonucleic Acid (cfDNA) Detected
    53
    35.6%
    58
    39.5%
    Other (Sample failed analysis, uninformative, or not analyzed.)
    4
    2.7%
    2
    1.4%
    ≥1 ALK Mutation Detected
    3
    2%
    5
    3.4%
    No ALK Mutation Detected
    45
    30.2%
    42
    28.6%
    No Circulating Free Deoxyribonucleic Acid (cfDNA) Detected
    52
    34.9%
    35
    23.8%
    Other (Sample failed analysis, uninformative, or not analyzed.)
    3
    2%
    2
    1.4%
    28. Secondary Outcome
    Title Number of Participant With ALK Fusion Variant in Plasma Circulating Nucleic Acid (CNA) Analysis at Screening, Cycle 2 Day 1 and Cycle 7 Day 1
    Description The analysis of ALK fusion variant in plasma CNA was performed by NGS and the number of participants with fusion variants at Screening, Cycle 2 Day 1 and Cycle 7 Day 1 is presented here. In the table below, EML4-ALK is the abbreviation of echinoderm microtubule-associated protein-like 4 anaplastic lymphoma kinase.
    Time Frame at Screening, Cycle 2 Day 1 and Cycle 7 Day 1

    Outcome Measure Data

    Analysis Population Description
    "Number of Participants Analyzed" included all participants in the plasma CNA analysis population within each treatment group at screening. "Number Analyzed" included all participants in the plasma CNA analysis population within each treatment group at the specified visit.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    Measure Participants 130 125
    EML4-ALK Variant 1
    19
    12.8%
    25
    17%
    EML4-ALK Variant 2
    7
    4.7%
    2
    1.4%
    EML4-ALK Variant 3
    18
    12.1%
    21
    14.3%
    EML4-ALK Other
    15
    10.1%
    9
    6.1%
    ALK Rearrangement Other
    2
    1.3%
    4
    2.7%
    ALK Rearrangement Not Detected
    32
    21.5%
    36
    24.5%
    No cfDNA Detected
    32
    21.5%
    25
    17%
    Other (Sample failed analysis, uninformative, or not analyzed.)
    5
    3.4%
    3
    2%
    EML4-ALK Variant 1
    1
    0.7%
    7
    4.8%
    EML4-ALK Variant 2
    2
    1.3%
    0
    0%
    EML4-ALK Variant 3
    2
    1.3%
    2
    1.4%
    EML4-ALK Other
    2
    1.3%
    1
    0.7%
    ALK Rearrangement Other
    0
    0%
    0
    0%
    ALK Rearrangement Not Detected
    61
    40.9%
    48
    32.7%
    No cfDNA Detected
    53
    35.6%
    58
    39.5%
    Other (Sample failed analysis, uninformative, or not analyzed.)
    4
    2.7%
    2
    1.4%
    EML4-ALK Variant 1
    0
    0%
    5
    3.4%
    EML4-ALK Variant 2
    0
    0%
    1
    0.7%
    EML4-ALK Variant 3
    0
    0%
    4
    2.7%
    EML4-ALK Other
    0
    0%
    1
    0.7%
    ALK Rearrangement Other
    0
    0%
    2
    1.4%
    ALK Rearrangement Not Detected
    48
    32.2%
    34
    23.1%
    No cfDNA Detected
    52
    34.9%
    35
    23.8%
    Other (Sample failed analysis, uninformative, or not analyzed.)
    3
    2%
    2
    1.4%

    Adverse Events

    Time Frame From the first dose of study treatment through end of study follow-up (infinite lag) or start of new anti-cancer therapy, whichever occurred first (assessed up to 33 months).
    Adverse Event Reporting Description The analysis of adverse events included all participants who received at least 1 dose of study drug. Participants were classified according to the treatment assigned at randomization. Totals number of participants at a higher level were not necessarily the sum of those at the lower levels since a participant may report two or more different adverse events within the higher level category.
    Arm/Group Title Lorlatinib Crizotinib
    Arm/Group Description Lorlatinib monotherapy at the recommended Phase 2 dose of 100 mg, was administered orally once daily (QD) at approximately the same time of the day on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first. Crizotinib monotherapy at the registered dose of 250 mg, was administered orally twice daily (BID) at approximately the same time in the morning and evening (12 hours apart) on a continuous daily dosing schedule. Each treatment cycle was defined as 28 days. Treatment continued until confirmation of RECIST version 1.1 defined disease progression assessed by the blinded independent central review (unless clinical benefit was still achievable), participant refusal, participant lost to follow-up, unacceptable toxicity, death, or the study was terminated by the sponsor, whichever occurred first.
    All Cause Mortality
    Lorlatinib Crizotinib
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 23/149 (15.4%) 28/142 (19.7%)
    Serious Adverse Events
    Lorlatinib Crizotinib
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 51/149 (34.2%) 39/142 (27.5%)
    Blood and lymphatic system disorders
    Anaemia 1/149 (0.7%) 0/142 (0%)
    Cardiac disorders
    Cardiac disorder 1/149 (0.7%) 0/142 (0%)
    Cardiac failure 2/149 (1.3%) 1/142 (0.7%)
    Cardiac failure acute 1/149 (0.7%) 0/142 (0%)
    Cardiac tamponade 1/149 (0.7%) 0/142 (0%)
    Left ventricular dysfunction 1/149 (0.7%) 0/142 (0%)
    Pericardial effusion 2/149 (1.3%) 1/142 (0.7%)
    Sinus node dysfunction 0/149 (0%) 1/142 (0.7%)
    Eye disorders
    Blindness cortical 0/149 (0%) 1/142 (0.7%)
    Retinal detachment 0/149 (0%) 1/142 (0.7%)
    Gastrointestinal disorders
    Constipation 0/149 (0%) 1/142 (0.7%)
    Diarrhoea 1/149 (0.7%) 0/142 (0%)
    Dyspepsia 0/149 (0%) 1/142 (0.7%)
    Oesophagitis 0/149 (0%) 1/142 (0.7%)
    Pancreatitis 1/149 (0.7%) 0/142 (0%)
    General disorders
    Death 1/149 (0.7%) 1/142 (0.7%)
    Disease progression 1/149 (0.7%) 1/142 (0.7%)
    Generalised oedema 1/149 (0.7%) 0/142 (0%)
    Pyrexia 3/149 (2%) 3/142 (2.1%)
    Hepatobiliary disorders
    Cholecystitis chronic 0/149 (0%) 1/142 (0.7%)
    Hepatic function abnormal 0/149 (0%) 1/142 (0.7%)
    Liver injury 0/149 (0%) 1/142 (0.7%)
    Immune system disorders
    Anaphylactic reaction 1/149 (0.7%) 0/142 (0%)
    Infections and infestations
    Bacterial infection 0/149 (0%) 1/142 (0.7%)
    Bronchitis 2/149 (1.3%) 0/142 (0%)
    Clostridium difficile colitis 0/149 (0%) 1/142 (0.7%)
    Gastroenteritis 0/149 (0%) 1/142 (0.7%)
    Haemophilus infection 1/149 (0.7%) 0/142 (0%)
    Lung abscess 1/149 (0.7%) 0/142 (0%)
    Pharyngeal abscess 0/149 (0%) 1/142 (0.7%)
    Pharyngitis 1/149 (0.7%) 0/142 (0%)
    Pneumonia 7/149 (4.7%) 5/142 (3.5%)
    Pneumonia cryptococcal 1/149 (0.7%) 0/142 (0%)
    Respiratory tract infection 1/149 (0.7%) 0/142 (0%)
    Sepsis 2/149 (1.3%) 0/142 (0%)
    Tuberculosis 0/149 (0%) 1/142 (0.7%)
    Upper respiratory tract infection 1/149 (0.7%) 1/142 (0.7%)
    Urinary tract infection 1/149 (0.7%) 0/142 (0%)
    Injury, poisoning and procedural complications
    Femoral neck fracture 0/149 (0%) 1/142 (0.7%)
    Femur fracture 0/149 (0%) 1/142 (0.7%)
    Fracture 1/149 (0.7%) 0/142 (0%)
    Hip fracture 1/149 (0.7%) 0/142 (0%)
    Lower limb fracture 1/149 (0.7%) 0/142 (0%)
    Multiple injuries 1/149 (0.7%) 0/142 (0%)
    Pelvic fracture 0/149 (0%) 1/142 (0.7%)
    Skin laceration 1/149 (0.7%) 0/142 (0%)
    Investigations
    Blood triglycerides increased 1/149 (0.7%) 0/142 (0%)
    Haemoglobin decreased 1/149 (0.7%) 0/142 (0%)
    Neutrophil count decreased 0/149 (0%) 1/142 (0.7%)
    Platelet count decreased 0/149 (0%) 1/142 (0.7%)
    Metabolism and nutrition disorders
    Diabetes mellitus 1/149 (0.7%) 0/142 (0%)
    Hypercholesterolaemia 1/149 (0.7%) 0/142 (0%)
    Hypertriglyceridaemia 1/149 (0.7%) 0/142 (0%)
    Hypocalcaemia 0/149 (0%) 1/142 (0.7%)
    Hyponatraemia 1/149 (0.7%) 1/142 (0.7%)
    Musculoskeletal and connective tissue disorders
    Back pain 2/149 (1.3%) 0/142 (0%)
    Muscular weakness 0/149 (0%) 1/142 (0.7%)
    Osteoarthritis 1/149 (0.7%) 0/142 (0%)
    Pain in extremity 1/149 (0.7%) 0/142 (0%)
    Rotator cuff syndrome 1/149 (0.7%) 0/142 (0%)
    Spondylitis 1/149 (0.7%) 0/142 (0%)
    Tenosynovitis 1/149 (0.7%) 0/142 (0%)
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Endometrial adenocarcinoma 1/149 (0.7%) 0/142 (0%)
    Follicle centre lymphoma, follicular grade I, II, III 1/149 (0.7%) 0/142 (0%)
    Lung neoplasm malignant 1/149 (0.7%) 0/142 (0%)
    Malignant neoplasm progression 0/149 (0%) 2/142 (1.4%)
    Neoplasm progression 0/149 (0%) 1/142 (0.7%)
    Nervous system disorders
    Carpal tunnel syndrome 1/149 (0.7%) 0/142 (0%)
    Cerebral infarction 0/149 (0%) 1/142 (0.7%)
    Cognitive disorder 1/149 (0.7%) 0/142 (0%)
    Ischaemic stroke 0/149 (0%) 1/142 (0.7%)
    Speech disorder 1/149 (0.7%) 0/142 (0%)
    Thalamus haemorrhage 1/149 (0.7%) 0/142 (0%)
    Psychiatric disorders
    Confusional state 1/149 (0.7%) 0/142 (0%)
    Delirium 1/149 (0.7%) 0/142 (0%)
    Intentional self-injury 1/149 (0.7%) 0/142 (0%)
    Renal and urinary disorders
    Renal failure 1/149 (0.7%) 0/142 (0%)
    Urinary retention 0/149 (0%) 1/142 (0.7%)
    Urinary tract disorder 1/149 (0.7%) 0/142 (0%)
    Respiratory, thoracic and mediastinal disorders
    Chronic obstructive pulmonary disease 1/149 (0.7%) 0/142 (0%)
    Dyspnoea 4/149 (2.7%) 0/142 (0%)
    Epistaxis 0/149 (0%) 1/142 (0.7%)
    Pleural effusion 2/149 (1.3%) 2/142 (1.4%)
    Pneumonitis 2/149 (1.3%) 2/142 (1.4%)
    Pneumothorax 1/149 (0.7%) 0/142 (0%)
    Pulmonary embolism 1/149 (0.7%) 2/142 (1.4%)
    Respiratory failure 4/149 (2.7%) 0/142 (0%)
    Surgical and medical procedures
    Radiotherapy to bone 1/149 (0.7%) 0/142 (0%)
    Vascular disorders
    Aortitis 0/149 (0%) 1/142 (0.7%)
    Hypertensive crisis 1/149 (0.7%) 0/142 (0%)
    Thrombosis 0/149 (0%) 1/142 (0.7%)
    Other (Not Including Serious) Adverse Events
    Lorlatinib Crizotinib
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 148/149 (99.3%) 140/142 (98.6%)
    Blood and lymphatic system disorders
    Anaemia 29/149 (19.5%) 11/142 (7.7%)
    Neutropenia 10/149 (6.7%) 21/142 (14.8%)
    Cardiac disorders
    Bradycardia 2/149 (1.3%) 17/142 (12%)
    Sinus bradycardia 4/149 (2.7%) 15/142 (10.6%)
    Ear and labyrinth disorders
    Tinnitus 9/149 (6%) 1/142 (0.7%)
    Eye disorders
    Photopsia 1/149 (0.7%) 24/142 (16.9%)
    Vision blurred 11/149 (7.4%) 4/142 (2.8%)
    Visual impairment 10/149 (6.7%) 16/142 (11.3%)
    Gastrointestinal disorders
    Abdominal distension 9/149 (6%) 3/142 (2.1%)
    Abdominal pain 7/149 (4.7%) 11/142 (7.7%)
    Constipation 26/149 (17.4%) 41/142 (28.9%)
    Diarrhoea 32/149 (21.5%) 74/142 (52.1%)
    Flatulence 11/149 (7.4%) 3/142 (2.1%)
    Nausea 22/149 (14.8%) 74/142 (52.1%)
    Vomiting 19/149 (12.8%) 55/142 (38.7%)
    General disorders
    Asthenia 20/149 (13.4%) 27/142 (19%)
    Chest pain 16/149 (10.7%) 20/142 (14.1%)
    Fatigue 11/149 (7.4%) 24/142 (16.9%)
    Oedema 16/149 (10.7%) 13/142 (9.2%)
    Oedema peripheral 65/149 (43.6%) 44/142 (31%)
    Pain 5/149 (3.4%) 8/142 (5.6%)
    Pyrexia 22/149 (14.8%) 16/142 (11.3%)
    Infections and infestations
    Bronchitis 9/149 (6%) 3/142 (2.1%)
    Nasopharyngitis 8/149 (5.4%) 10/142 (7%)
    Pneumonia 5/149 (3.4%) 8/142 (5.6%)
    Respiratory tract infection 12/149 (8.1%) 6/142 (4.2%)
    Upper respiratory tract infection 16/149 (10.7%) 11/142 (7.7%)
    Urinary tract infection 6/149 (4%) 10/142 (7%)
    Investigations
    Alanine aminotransferase increased 26/149 (17.4%) 48/142 (33.8%)
    Amylase increased 13/149 (8.7%) 16/142 (11.3%)
    Aspartate aminotransferase increased 21/149 (14.1%) 39/142 (27.5%)
    Blood alkaline phosphatase increased 7/149 (4.7%) 18/142 (12.7%)
    Blood cholesterol increased 59/149 (39.6%) 4/142 (2.8%)
    Blood creatine phosphokinase increased 16/149 (10.7%) 24/142 (16.9%)
    Blood creatinine increased 9/149 (6%) 19/142 (13.4%)
    Blood lactate dehydrogenase increased 3/149 (2%) 15/142 (10.6%)
    Blood triglycerides increased 19/149 (12.8%) 2/142 (1.4%)
    Electrocardiogram QT prolonged 5/149 (3.4%) 8/142 (5.6%)
    Gamma-glutamyltransferase increased 22/149 (14.8%) 22/142 (15.5%)
    Lipase increased 14/149 (9.4%) 17/142 (12%)
    Neutrophil count decreased 3/149 (2%) 16/142 (11.3%)
    Weight decreased 4/149 (2.7%) 10/142 (7%)
    Weight increased 57/149 (38.3%) 18/142 (12.7%)
    White blood cell count decreased 1/149 (0.7%) 11/142 (7.7%)
    Metabolism and nutrition disorders
    Decreased appetite 5/149 (3.4%) 35/142 (24.6%)
    Dyslipidaemia 9/149 (6%) 0/142 (0%)
    Hypercholesterolaemia 53/149 (35.6%) 1/142 (0.7%)
    Hyperglycaemia 15/149 (10.1%) 5/142 (3.5%)
    Hyperlipidaemia 16/149 (10.7%) 0/142 (0%)
    Hypertriglyceridaemia 78/149 (52.3%) 6/142 (4.2%)
    Hyperuricaemia 12/149 (8.1%) 4/142 (2.8%)
    Hypoalbuminaemia 8/149 (5.4%) 18/142 (12.7%)
    Hypocalcaemia 2/149 (1.3%) 8/142 (5.6%)
    Hypokalaemia 9/149 (6%) 4/142 (2.8%)
    Musculoskeletal and connective tissue disorders
    Arthralgia 28/149 (18.8%) 16/142 (11.3%)
    Back pain 22/149 (14.8%) 16/142 (11.3%)
    Musculoskeletal pain 8/149 (5.4%) 5/142 (3.5%)
    Myalgia 16/149 (10.7%) 5/142 (3.5%)
    Pain in extremity 25/149 (16.8%) 12/142 (8.5%)
    Nervous system disorders
    Dizziness 16/149 (10.7%) 20/142 (14.1%)
    Dysgeusia 8/149 (5.4%) 23/142 (16.2%)
    Headache 25/149 (16.8%) 25/142 (17.6%)
    Hypoaesthesia 8/149 (5.4%) 7/142 (4.9%)
    Memory impairment 13/149 (8.7%) 3/142 (2.1%)
    Neuropathy peripheral 13/149 (8.7%) 0/142 (0%)
    Paraesthesia 18/149 (12.1%) 7/142 (4.9%)
    Peripheral sensory neuropathy 10/149 (6.7%) 1/142 (0.7%)
    Psychiatric disorders
    Anxiety 10/149 (6.7%) 3/142 (2.1%)
    Insomnia 14/149 (9.4%) 13/142 (9.2%)
    Renal and urinary disorders
    Proteinuria 9/149 (6%) 1/142 (0.7%)
    Respiratory, thoracic and mediastinal disorders
    Cough 24/149 (16.1%) 26/142 (18.3%)
    Dyspnoea 28/149 (18.8%) 23/142 (16.2%)
    Oropharyngeal pain 9/149 (6%) 8/142 (5.6%)
    Productive cough 12/149 (8.1%) 4/142 (2.8%)
    Skin and subcutaneous tissue disorders
    Hyperhidrosis 8/149 (5.4%) 0/142 (0%)
    Rash 15/149 (10.1%) 11/142 (7.7%)
    Vascular disorders
    Hypertension 27/149 (18.1%) 3/142 (2.1%)

    Limitations/Caveats

    [Not Specified]

    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:
    NCT03052608
    Other Study ID Numbers:
    • B7461006
    • 2016-003315-35
    First Posted:
    Feb 14, 2017
    Last Update Posted:
    May 31, 2022
    Last Verified:
    May 1, 2022