ClinicalTrials.gov

History of Changes for Study: NCT03915951
An Open-label Study of Encorafenib + Binimetinib in Patients With BRAFV600-mutant Non-small Cell Lung Cancer
Latest version (submitted April 12, 2024) on ClinicalTrials.gov
  • A study version is represented by a row in the table.
  • Select two study versions to compare. One each from columns A and B.
  • Choose either the "Merged" or "Side-by-Side" comparison format to specify how the two study versions are to be displayed. The Side-by-Side format only applies to the Protocol section of the study.
  • Click "Compare" to do the comparison and show the differences.
  • Select a version's Submitted Date link to see a rendering of the study for that version.
  • The yellow A/B choices in the table indicate the study versions currently compared below. A yellow table row indicates the study version currently being viewed.
  • Hover over the "Recruitment Status" to see how the study's recruitment status changed.
  • Study edits or deletions are displayed in red.
  • Study additions are displayed in green.
Study Record Versions
Version A B Submitted Date Changes
1 April 12, 2019 None (earliest Version on record)
2 June 28, 2019 Recruitment Status, Study Status, Contacts/Locations and Oversight
3 September 30, 2019 Study Status and Contacts/Locations
4 November 5, 2019 Contacts/Locations and Study Status
5 June 25, 2020 Contacts/Locations, Study Identification, Sponsor/Collaborators, Study Status and IPDSharing
6 August 16, 2020 Contacts/Locations, Study Status, References and Study Identification
7 September 15, 2020 Contacts/Locations, Study Status, Oversight and Study Identification
8 November 4, 2020 Contacts/Locations, Study Status and Study Design
9 November 14, 2020 Contacts/Locations, Study Design and Study Status
10 February 7, 2021 Contacts/Locations, Study Status and Study Design
11 April 12, 2021 Contacts/Locations, Study Status and Study Identification
12 May 11, 2021 Contacts/Locations and Study Status
13 May 25, 2021 Contacts/Locations, Outcome Measures, Study Status and Study Identification
14 July 14, 2021 Contacts/Locations and Study Status
15 August 13, 2021 Study Status and Contacts/Locations
16 September 27, 2021 Study Status, Contacts/Locations and Oversight
17 November 17, 2021 Study Status, Eligibility, Outcome Measures and Conditions
18 December 10, 2021 Study Status and Contacts/Locations
19 January 11, 2022 Contacts/Locations and Study Status
20 February 25, 2022 Study Status and Contacts/Locations
21 April 18, 2022 Contacts/Locations, Study Status and Study Design
22 May 23, 2022 Study Status and Contacts/Locations
23 June 13, 2022 Recruitment Status, Study Status, Contacts/Locations and Study Design
24 August 12, 2022 Contacts/Locations and Study Status
25 October 5, 2022 Study Status, Contacts/Locations and Study Design
26 November 16, 2022 Contacts/Locations and Study Status
27 December 29, 2022 Study Status and Contacts/Locations
28 February 16, 2023 Study Status and Contacts/Locations
29 March 16, 2023 Study Status and Contacts/Locations
30 April 17, 2023 Study Status
31 July 10, 2023 Study Status and Contacts/Locations
32 September 18, 2023
Quality Control Review has not concluded Returned: October 10, 2023
Outcome Measures, Study Status, Document Section and Contacts/Locations
33 October 26, 2023 Study Status, Outcome Measures
34 March 20, 2024 Study Status and Contacts/Locations
35 April 12, 2024 Study Status and Contacts/Locations
Comparison Format:

Scroll up to access the controls

Study NCT03915951
Submitted Date:  September 18, 2023 (v32)
Quality Control Review Has Not Concluded

Note: The results information displayed below has not completed the quality control (QC) review process. ClinicalTrials.gov must post results information for applicable clinical trials (ACTs) within 30 days of submission, even if the submission has not completed the QC review process. The study sponsor or investigator is responsible for ensuring the results information meets the QC review criteria.

This submission includes brief standardized QC review comments added by the National Library of Medicine (NLM). These comments indicate the location of apparent errors, deficiencies, or inconsistencies. For more information, see the Final Rule (42 CFR Part 11) Information page.


Open or close this module Study Identification
Unique Protocol ID: ARRAY-818-202
Brief Title: An Open-label Study of Encorafenib + Binimetinib in Patients With BRAFV600-mutant Non-small Cell Lung Cancer
Official Title: A Phase 2, Open-label Study of Encorafenib + Binimetinib in Patients With BRAFV600-mutant Non-small Cell Lung Cancer
Secondary IDs: C4221008 [Alias Study Number]
2019-000417-37 [EudraCT Number]
Open or close this module Study Status
Record Verification: September 2023
Overall Status: Active, not recruiting
Study Start: June 4, 2019
Primary Completion: September 22, 2022 [Actual]
Study Completion: December 16, 2024 [Anticipated]
First Submitted: April 12, 2019
First Submitted that
Met QC Criteria:
April 12, 2019
First Posted: April 16, 2019 [Actual]
Last Update Submitted that
Met QC Criteria:
Last Update Posted: October 11, 2023 [Actual]
Open or close this module Sponsor/Collaborators
Sponsor: Pfizer
Responsible Party: Sponsor
Collaborators:
Open or close this module Oversight
U.S. FDA-regulated Drug: Yes
U.S. FDA-regulated Device: No
Data Monitoring: No
Open or close this module Study Description
Brief Summary: This is an open-label, multicenter, non-randomized, Phase 2 study to determine the safety, tolerability and efficacy of encorafenib given in combination with binimetinib in patients with BRAFV600E-mutant metastatic non-small cell lung cancer (NSCLC). Patients who are either treatment-naïve, OR who have received 1) first-line treatment with standard platinum-based chemotherapy, OR 2) first-line treatment with an anti-programmed cell death protein 1 (PD-1)/programmed cell death protein ligand 1 (PD-L1) inhibitor given alone or in combination with platinum-based chemotherapy will be enrolled.
Detailed Description:
Open or close this module Conditions
Conditions: Non-small Cell Lung Cancer
Keywords: lung cancer
cancer
non-small cell lung cancer
non small cell lung cancer
NSCLC
Encorafenib
Binimetinib
Phase 2
Open Label
BRAF mutation
BRAF V600E
Array
Stage IV NSCLC
Metastatic NSCLC
PDL1
PD-LI
Immunotherapy
First line platinum based chemotherapy
BRAF inhibitor
V600
Open or close this module Study Design
Study Type: Interventional
Primary Purpose: Treatment
Study Phase: Phase 2
Interventional Study Model: Single Group Assignment
Number of Arms: 1
Masking: None (Open Label)
Allocation: N/A
Enrollment: 98 [Actual]
Open or close this module Arms and Interventions
Arms Assigned Interventions
Experimental: Treatment Period

Study treatment with encorafenib and binimetinib will be self-administered orally without regard to food.

Patients will receive the following per 28-day (± 3 days) cycle:

  • Encorafenib: 450 mg (6 × 75 mg capsule) once daily (QD)
  • Binimetinib: 45 mg (3 × 15 mg tablet) twice daily (BID)
Drug: encorafenib
self-administered orally
Drug: binimetinib
self-administered orally
Open or close this module Outcome Measures
[See Results Section.]
Primary Outcome Measures:
1. Percentage of Participants With Confirmed Objective Response (OR) as Determined by Independent Radiology Review (IRR)
[ Time Frame: From date of the first dose of study intervention until documented progressive disease (PD) or start of new anticancer therapy (up to 36 months) ]

Objective Response Rate (ORR) was defined as the percentage of participants who had achieved a confirmed best overall response (Complete Response [CR] or Partial Response [PR]) per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. Both CR and PR must be confirmed by repeat assessments performed no less than 4 weeks after the criteria for response were first met. CR was defined by the disappearance of all non-lymph node target lesions (where all target lesions were recorded with a length of 0 mm, and any pathological lymph nodes [recorded as target lesion] must have reduction in short axis to <10 mm) and complete disappearance of all non-target lesions (where all non-target lesions were marked "Absent", and all lymph nodes must be non-pathological in size [<10 mm in short axis]). PR was defined by a 30% or more decrease in sum of diameters (SOD) of target lesions, taking as reference the baseline SOD. ORR was calculated with the exact 2-sided Clopper-Pearson 95% CI.
Secondary Outcome Measures:
1. Percentage of Participants With Confirmed Objective Response (OR) by Investigator Assessment
[ Time Frame: From date of the first dose of study intervention until documented PD or start of new anticancer therapy (up to 36 months) ]

ORR was defined as the percentage of participants who had achieved a confirmed best overall response (CR or PR) per RECIST version 1.1. Both CR and PR must be confirmed by repeat assessments performed no less than 4 weeks after the criteria for response were first met. CR was defined by the disappearance of all non-lymph node target lesions (where all target lesions were recorded with a length of 0 mm, and any pathological lymph nodes [recorded as target lesion] must have reduction in short axis to <10 mm) and the complete disappearance of all non-target lesions (where all non-target lesions were marked "Absent", and all lymph nodes must be non-pathological in size [<10 mm in short axis]). PR was defined by a 30% or more decrease in SOD of target lesions, taking as reference the baseline SOD. ORR according to derived investigator assessment was calculated with the exact 2-sided Clopper-Pearson 95% CI.
2. Duration of Response (DoR) by IRR and Investigator Assessments
[ Time Frame: From the date of the first confirmed documented response (CR or PR) to the earliest date of disease progression or death due to any cause (up to 36 months) ]

DoR, based on IRR and Investigator assessments was defined as the time from the date of the first documented confirmed response (CR or PR) (by IRR and by Investigator, respectively) to the earliest date of disease progression, as determined by Investigator review of radiographic disease assessments and IRR per RECIST version 1.1, or death due to any cause. If a participant with a CR or PR had neither progressed nor died at the time of the analysis cutoff or at the start of any new anticancer therapy, the participant was censored at the date of last adequate tumor assessment. CR was defined by the disappearance of all non-lymph node target lesions and complete disappearance of all non-target lesions. PR was defined by a 30% or more decrease in SOD of target lesions, taking as reference the baseline SOD. DoR was calculated for participants who had achieved a confirmed overall response (CR or PR). The estimate of the DoR survival function was constructed using the Kaplan-Meier method.
3. Disease Control Rate (DCR) by IRR and Investigator Assessments
[ Time Frame: After 24 Weeks (≥168 days) from the date of first dose of study intervention ]

DCR was defined as the percentage of participants who had achieved a confirmed overall response of CR, PR or stable disease (SD), as determined by Investigator review of radiographic disease assessments and IRR per RECIST version 1.1 after 24 weeks (≥168 days) from the date of first dose of study intervention. CR was defined by the disappearance of all non-lymph node target lesions and complete disappearance of all non-target lesions. PR was defined by a 30% or more decrease in SOD of target lesions, taking as reference the baseline SOD. SD was assigned when neither sufficient shrinkage to qualify for CR or PR, nor sufficient increase to qualify for PD was observed, taking as reference the nadir. DCR was calculated along with the exact 2-sided Clopper-Pearson 95% CI.
4. Progression-free Survival (PFS) by IRR and Investigator Assessments
[ Time Frame: From the date of first dose of study drug to the earliest date of disease progression, or death due to any cause (up to 36 months) ]

PFS was defined as the time from the date of first dose of study intervention to the earliest date of disease progression, as determined by IRR and Investigator review of radiographic disease assessments per RECIST version 1.1, or death due to any cause, whichever occurred first. PD was defined by a 20% or more increase in the SOD of target lesions relative to nadir (smallest SOD considering baseline and all assessments prior to the time point under evaluation), with a minimum absolute increase of 5 mm relative to nadir; PD was assigned if any non-target lesion was marked "unequivocal progression". If a participant had not had a PFS event at the time of the analysis cutoff or at the start of any new anticancer therapy, PFS was censored at the date of last adequate tumor assessment. PFS (months) = [(date of event or censoring - date of first dose) +1]/30.4375. The survival distribution function for PFS was estimated using the Kaplan-Meier method.
5. Time to Response (TTR) by IRR and Investigator Assessments
[ Time Frame: From the date of first dose to the first documentation of confirmed objective response (CR or PR) (up to 36 months) ]

TTR based on IRR and Investigator assessments was defined, for participants with an objective response, as the time, in months, from the date of first dose to the first documentation of objective response (CR or PR) which was subsequently confirmed (by IRR and by Investigator, respectively). TTR was calculated for the subgroup of participants with a confirmed objective tumor response.
6. Kaplan-Meier Estimates of Overall Survival (OS)
[ Time Frame: The time from the date of first dose of study intervention to the date of death due to any cause (up to 36 months) ]

OS was defined as the time from the date of first dose of study intervention to the date of death due to any cause. If a death had not been observed by the date of the analysis cutoff, OS was censored at the date of last contact. The survival distribution function for OS was estimated using the Kaplan-Meier method.
7. Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
[ Time Frame: From the time the participant provided informed consent, through and including a minimum of 30 calendar days, after the last administration of the study intervention, and the safety follow-up visit (30 days [±7 days] after the EOT visit) (up to 36 months) ]

TEAE was a treatment-emergent adverse event that occurred during the on-treatment period. The on-treatment period was defined as the time from the first dose date of study intervention to the last dose of study drug administration date (when both drugs were permanently discontinued) +30 days or the earliest date of subsequent anti-cancer drug therapy minus 1 day, whichever occurred first. Relatedness to study intervention was determined by the investigator. The investigator made an assessment of intensity for each AE reported during the study according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.03: Grade 3 events = severe AEs; Grade 4 events = life-threatening consequences, urgent intervention indicated; Grade 5 events = death related to AEs.
8. Number of Participants With Shifts From Grade ≤2 at Baseline to Grade 3 or 4 at Post-baseline in Hematology Laboratory Test Values Based on CTCAE Grade
[ Time Frame: Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Day 1 ±3 Days in Subsequent Cycles, End of Treatment (EOT) ±3 Days and Safety Follow-up Visit (30 Days [±7 Days] After the EOT Visit) (Up to 36 Months) ]

Blood and urine samples for the laboratory tests. A central laboratory will perform all clinical laboratory assessments. Baseline was the last available assessment performed prior to the study intervention start date/time.
9. Number of Participants With Shifts From Grade ≤2 at Baseline to Grade 3 or 4 at Post-baseline in Chemistry Laboratory Test Values Based on CTCAE Grade
[ Time Frame: Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Day 1 ±3 Days in Subsequent Cycles, EOT ±3 Days and Safety Follow-up Visit (30 Days [±7 Days] After the EOT Visit) (Up to 36 Months) ]

Blood and urine samples for the laboratory tests. A central laboratory will perform all clinical laboratory assessments. Baseline was the last available assessment performed prior to the study intervention start date/time.
10. Number of Participants With Notable Abnormal Vital Signs
[ Time Frame: Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Day 1 ±3 Days of Subsequent Cycles, EOT ±3 Days, and Safety Follow-up Visit (30 Days [±7 Days] After the EOT Visit) (Up to 36 Months) ]

Vital sign measurements were taken before blood collection for laboratory tests or at least 30 minutes after blood collection for laboratory tests, and were measured per institutional standards. Vital sign assessments included temperature, pulse rate, respiratory rate, and blood pressure (assessed in a recumbent, semi recumbent, or sitting position).

The criteria of notably abnormal vital signs are listed below:

Systolic blood pressure (mmHg): high: ≥160 mmHg with increase from baseline of ≥20 mmHg; low: ≤90 mmHg with decrease from baseline of ≥20 mmHg.

Diastolic blood pressure (mmHg): high: ≥100 mmHg with increase from baseline of ≥15 mmHg; low: ≤50 mmHg with decrease from baseline of ≥15 mmHg.

Pulse rate (bpm): high: ≥120 bpm with increase from baseline of ≥15 bpm; low: ≤50 bpm with decrease from baseline of ≥15 bpm.

Weight (kg): high: ≥10% increase from baseline; low: ≥20% decrease from baseline.

Temperature (°C): high: ≥37.5 °C; low: ≤36 °C.

11. Number of Participants With Notable ECG (QTcF) Values
[ Time Frame: Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Every 12 Weeks ±7 Days, and EOT ±3 Days (Up to 36 Months) ]

The QTcF increase from baseline >30/60 msec and new QTcF >450/480/500 msec were defined as clinically notable ECG criteria. Triplicate ECG measurements were obtained. For new abnormal post-baseline values, the table below presents the number of participants with both non-missing baseline and post-baseline values, and baseline values not meeting the criteria. For abnormal changes from baseline, the table below presents the number of participants with both non-missing baseline and post-baseline evaluations. Baseline was defined as the average of the machine-read triplicate ECG measurements taken pre-dose on Day 1. Change from baseline was post-baseline - baseline values.
12. Number of Participants With the Worst Post-baseline Left Ventricular Ejection Fraction (LVEF) Values Based on CTCAE Grade
[ Time Frame: Screening, Cycle 2 Day 1 ±3 Days, Every 12 Weeks ±7 Days, and EOT ±3 Days (Up to 36 Months) ]

Cardiac ejection fraction was assessed by transthoracic echocardiogram (ECHO) or multigated acquisition (MUGA). Participants who developed signs/symptoms of congestive heart failure (CHF) at any point during the study were required to have an evaluation of LVEF measurements by ECHO or MUGA and were monitored per institutional guidelines.

Participants were considered as having a LVEF abnormality if the worst post-value was CTCAE Grade 2, 3 or 4 according to the following classification:

Grade 0: Non-missing value below Grade 2. Grade 2: LVEF between 40% and 50%, inclusive, or absolute change from baseline between -10% and < -20%.

Grade 3: LVEF between 20% and 39%, inclusive, or absolute change from baseline ≤ -20%.

Grade 4: LVEF lower than 20%. Baseline was defined as the last available and valid assessment before or on the start date of study intervention.

Open or close this module Eligibility
Minimum Age: 18 Years
Maximum Age:
Sex: All
Gender Based:
Accepts Healthy Volunteers: No
Criteria:

Key Inclusion Criteria:

  • Histologically confirmed diagnosis of non-small cell lung cancer (NSCLC) that is currently Stage IV.
  • Presence of a BRAFV600E mutation in lung cancer tissue as determined by a local laboratory assay or the presence of other BRAFV600 mutations other than V600E (i.e. K or D) will be considered
  • Patients who are either treatment-naïve (e.g., no prior systemic therapy for advanced/metastatic disease), OR who have received 1) first-line platinum-based chemotherapy OR 2) first-line treatment with an anti-programmed cell death protein 1 (PD-1)/ programmed cell death protein ligand 1(PD-L1) inhibitor given alone or in combination with platinum-based chemotherapy.
  • Presence of measurable disease based on Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1).
  • Eastern Cooperation Oncology Group (ECOG) performance status of 0 or 1.
  • Adequate bone marrow function characterized by the following at screening:
    • absolute neutrophil count (ANC) ≥ 1.5 × 10⁹/L;
    • Platelets ≥ 100 × 10⁹/L;
    • Hemoglobin ≥ 8.5 g/dL (with or without blood transfusions).
  • Adequate hepatic and renal function characterized by the following at screening:
    • Total bilirubin ≤ 1.5 × upper limit of normal (ULN)
    • alanine aminotransferase (ALT) and aspartate aminotransferase (AST) ≤ 2.5 × ULN, or ≤ 5 × ULN in presence of liver metastases; Serum creatinine ≤ 1.5 × ULN; or calculated creatinine clearance ≥ 50 mL/min by Cockcroft-Gault formula; or estimated glomerular filtration rate > 50 mL/min/1.73m².

Key Exclusion Criteria:

  • Patients who have documentation of any of the following:
    • epidermal growth factor receptor (EGFR) mutation
    • anaplastic lymphoma kinase (ALK) fusion oncogene or
    • ROS1 rearrangement
  • Patients who have received more than 1 prior line of systemic therapy in the advanced/metastatic setting.
  • Previous treatment with any BRAF inhibitor (e.g., dabrafenib, vemurafenib, XL281/BMS-908662, etc.), or any mitogen-activated protein kinase (MEK) inhibitor (e.g., trametinib, cobimetinib, selumetinib, RDEA119, etc.) prior to screening and enrollment.
  • Impaired cardiovascular function or clinically significant cardiovascular diseases
  • History of thromboembolic or cerebrovascular events ≤ 12 weeks prior to the first dose of study treatment. Examples include transient ischemic attacks, cerebrovascular accidents, hemodynamically significant (i.e. massive or sub-massive) deep vein thrombosis or pulmonary emboli.
  • History or current evidence of retinal vein occlusion (RVO) or current risk factors for RVO (e.g., uncontrolled glaucoma or ocular hypertension, history of hyperviscosity or hypercoagulability syndromes); history of retinal degenerative disease.
  • Concurrent neuromuscular disorder that is associated with the potential of elevated creatine (phospho)kinase (CK) (e.g., inflammatory myopathies, muscular dystrophy, amyotrophic lateral sclerosis, spinal muscular atrophy).
  • Patients with symptomatic brain metastasis, leptomeningeal disease or other active central nervous system (CNS) metastases are not eligible.
Open or close this module Contacts/Locations
Study Officials: Pfizer CT.gov Call Center
Study Director
Pfizer
Locations: United States, Arizona
Yuma Regional Medical Center Cancer Center
Yuma, Arizona, United States, 85364
Yuma Regional Medical Center Ophthalmology
Yuma, Arizona, United States, 85364
Yuma Regional Medical Center
Yuma, Arizona, United States, 85364
United States, California
UCLA Stein Eye Center Santa Monica (OPH)
Santa Monica, California, United States, 90403
UCLA Hematology/Oncology
Santa Monica, California, United States, 90404
United States, Florida
Florida Cancer Specialist
Altamonte Springs, Florida, United States, 32701
Florida Cancer Specialists
Bonita Springs, Florida, United States, 34135
Florida Cancer Specialist
Brandon, Florida, United States, 33511
Florida Cancer Specialists
Cape Coral, Florida, United States, 33909
Florida Cancer Specialist
Clearwater, Florida, United States, 33761
Florida Cancer Specialists
Fleming Island, Florida, United States, 32003
Florida Cancer Specialists
Fort Myers, Florida, United States, 33905
Florida Cancer Specialists
Fort Myers, Florida, United States, 33908
Florida Cancer Specialist
Gainesville, Florida, United States, 32605
Florida Cancer Specialists
Largo, Florida, United States, 33770
Florida Cancer Specialist
Lecanto, Florida, United States, 34461
Florida Cancer Specialists
Naples, Florida, United States, 34102
Florida Cancer Specialist
Ocala, Florida, United States, 34474
Florida Cancer Specialist
Orange City, Florida, United States, 32763
Florida Cancer Specialist
Orlando, Florida, United States, 32806
Florida Cancer Specialists
Port Charlotte, Florida, United States, 33980
Florida Cancer Specialist
Saint Petersburg, Florida, United States, 33705
Florida Cancer Specialists
Sarasota, Florida, United States, 34232
Florida Cancer Specialists
Sarasota, Florida, United States, 34236
Florida Cancer Specialists
Spring Hill, Florida, United States, 34608
Florida Cancer Specialists PAN - SCRI - PPDS
Tallahassee, Florida, United States, 32308
Florida Cancer Specialist
Tampa, Florida, United States, 33607
Florida Cancer Specialist
Tavares, Florida, United States, 32778
Florida Cancer Specialist
The Villages, Florida, United States, 32159
Florida Cancer Specialist
Trinity, Florida, United States, 34655
Florida Cancer Specialists
Venice, Florida, United States, 34285
Florida Cancer Specialists
Venice, Florida, United States, 34292
Florida Cancer Specialists
Winter Park, Florida, United States, 32792
United States, Georgia
Winship Cancer Institute @ Emory University Hospital Midtown
Atlanta, Georgia, United States, 30308
Emory University Hospital
Atlanta, Georgia, United States, 30322
Winship Cancer Institute, Emory University
Atlanta, Georgia, United States, 30322
Winship Cancer Institute @ Emory Saint Joseph's Hospital
Atlanta, Georgia, United States, 30342
Winship Cancer Institute @ Emory Johns Creek Hospital
Johns Creek, Georgia, United States, 30097
United States, Kansas
MidAmerica Division, Inc. c/o Menorah Medical Center
Overland Park, Kansas, United States, 66209
United States, Maryland
Johns Hopkins Bayview Medical Center
Baltimore, Maryland, United States, 21224
Johns Hopkins Sidney Kimmel Comprehensive Cancer Center
Baltimore, Maryland, United States, 21231
Johns Hopkins Sidney Kimmel Comprehensive Cancer Center
Baltimore, Maryland, United States, 21287
The Johns Hopkins Wilmer Eye Institute
Baltimore, Maryland, United States, 21287
United States, Massachusetts
Massachusetts General Hospital
Boston, Massachusetts, United States, 02114
Ophthalmic Consultants of Boston (OCB)
Boston, Massachusetts, United States, 02114
Ophthalmic Consultants of Boston Inc
Boston, Massachusetts, United States, 02114
Beth Israel Deaconess Medical Center
Boston, Massachusetts, United States, 02115
Brigham and Women's Hospital
Boston, Massachusetts, United States, 02115
Dana Farber Cancer Institute
Boston, Massachusetts, United States, 02115
Beth Israel Deaconess Medical Center - 330 Brookline Ave
Boston, Massachusetts, United States, 02215-5400
Beth Israel Deaconess Medical Center/East
Boston, Massachusetts, United States, 02215
Dana-Farber Cancer Institute - Chestnut Hill
Newton, Massachusetts, United States, 02459
United States, Missouri
Siteman Cancer Center - West County
Creve Coeur, Missouri, United States, 63141
MidAmerica Division, Inc. c/o Centerpoint Medical Center
Independence, Missouri, United States, 64057
MidAmerica Division, Inc., c/o Research Medical Center
Kansas City, Missouri, United States, 64132
Barnes-Jewish Hospital
Saint Louis, Missouri, United States, 63110
Washington University School of Medicine
Saint Louis, Missouri, United States, 63110
Siteman Cancer Center - South County
Saint Louis, Missouri, United States, 63129
Siteman Cancer Center - St Peters
Saint Peters, Missouri, United States, 63376
United States, New Jersey
Memorial Sloan Kettering Cancer Center Basking Ridge
Basking Ridge, New Jersey, United States, 07920
Hackensack University Medical Center
Hackensack, New Jersey, United States, 07601
Metropolitan Eye Care
Paramus, New Jersey, United States, 07652
United States, New York
Memorial Sloan Kettering Cancer Center David H Koch Center for Cancer Care
New York, New York, United States, 10021
Weill Cornell Eye Associates
New York, New York, United States, 10021
Weill Cornell Medical College - New York Presbyterian Hospital
New York, New York, United States, 10021
Memorial Sloan Kettering Cancer Center
New York, New York, United States, 10065
Weill Cornell Medical College - New York Presbyterian Hospital
New York, New York, United States, 10065
United States, North Carolina
Duke Cancer Center
Durham, North Carolina, United States, 27710
United States, Ohio
Ohio State CarePoint East
Columbus, Ohio, United States, 43203
The Ohio State University Hospital
Columbus, Ohio, United States, 43210
The Ohio State University James Cancer Hospital
Columbus, Ohio, United States, 43210
The Ohio State University Medical Center - Thoracic Oncology Clinic
Columbus, Ohio, United States, 43210
Ohio State Eye and Ear Institute
Columbus, Ohio, United States, 43212
Stefanie Spielman Comprehensive Breast Cancer
Columbus, Ohio, United States, 43212
Martha Morehouse Medical Plaza
Columbus, Ohio, United States, 43221
Ohio State CarePoint Gahanna
Gahanna, Ohio, United States, 43230
Ohio State Outpatient Care Lewis Center
Lewis Center, Ohio, United States, 43035
United States, Oregon
Providence Cancer Institute Clackamas Clinic
Clackamas, Oregon, United States, 97015
Providence Cancer Institute Newberg Clinic
Newberg, Oregon, United States, 97132
Providence Cancer Institute, Franz Clinic
Portland, Oregon, United States, 97213
Providence Oncology and Hematology Care Clinic - Westside
Portland, Oregon, United States, 97225
United States, Pennsylvania
UPMC Hillman Cancer Center Erie
Erie, Pennsylvania, United States, 16505
UPMC Hillman Cancer Center - Arnold Palmer - Mt View
Greensburg, Pennsylvania, United States, 15601
UPMC Hillman Cancer Center Monroeville
Monroeville, Pennsylvania, United States, 15146
UPMC Eye Center, Eye and Ear Institute
Pittsburgh, Pennsylvania, United States, 15213-2548
UPMC Hillman Cancer Center
Pittsburgh, Pennsylvania, United States, 15232
UPMC Hillman Cancer Center - Passavant (HOA)
Pittsburgh, Pennsylvania, United States, 15237
UPMC Hillman Cancer Center - Passavant (OHA)
Pittsburgh, Pennsylvania, United States, 15237
UPMC Hillman Cancer Center - Upper St. Clair
Pittsburgh, Pennsylvania, United States, 15243
Weaver Eye Associates
York, Pennsylvania, United States, 17402
WellSpan Oncology Research
York, Pennsylvania, United States, 17403
United States, Tennessee
Tennessee Oncology, PLLC
Chattanooga, Tennessee, United States, 37404
Tennessee Oncology, PLLC
Cleveland, Tennessee, United States, 37311
Tennessee Oncology, PLLC
Dickson, Tennessee, United States, 37055
Tennessee Oncology, PLLC
Franklin, Tennessee, United States, 37067
Tennessee Oncology, PLLC
Gallatin, Tennessee, United States, 37066
Tennessee Oncology, PLLC
Hendersonville, Tennessee, United States, 37075
Tennessee Oncology, PLLC
Hermitage, Tennessee, United States, 37076
Tennessee Oncology, PLLC
Lebanon, Tennessee, United States, 37090
Tennessee Oncology, PLLC
Murfreesboro, Tennessee, United States, 37129
Tennessee Oncology, PLLC
Nashville, Tennessee, United States, 37203
Tennessee Oncology, PLLC
Nashville, Tennessee, United States, 37205
Tennessee Oncology, PLLC
Nashville, Tennessee, United States, 37207
Tennessee Oncology, PLLC
Nashville, Tennessee, United States, 37211
Tennessee Oncology, PLLC
Shelbyville, Tennessee, United States, 37160
Tennessee Oncology, PLLC
Smyrna, Tennessee, United States, 37167
United States, Texas
Houston Eye Associates - Gramercy Location
Houston, Texas, United States, 77008
The University of Texas MD Anderson Cancer Center
Houston, Texas, United States, 77030-4009
Millennium Research & Clinical Development
Houston, Texas, United States, 77090-3063
United States, Washington
Eye associates Northwest
Seattle, Washington, United States, 98104
Seattle Cancer Care Alliance
Seattle, Washington, United States, 98109
Italy
UO Dermatologia Centro Tumori Cutanei (CTC)- Azienda Ospedaliero
Bologna, Italy, 40138
UO di Oftalmologia- Azienda Ospedaliero Universitaria Di Bologna
Bologna, Italy, 40138
UO Farmacia Clinica- Azienda Ospedaliero Universitaria Di Bologna
Bologna, Italy, 40138
UO Medicina Borghi- Azienda Ospedaliero Universitaria Di Bologna
Bologna, Italy, 40138
UO Radiologia Golfieri- Azienda Ospedaliero Universitaria Di Bologna - Policlinico S Orsola Malpighi
Bologna, Italy, 40138
UOC di Anatomia Patologica- Azienda Ospedaliero Universitaria Di Bo logna
Bologna, Italy, 40138
IRCCS Ospedale San Raffaele
Milan, Italy, 20132
Clinica Oculistica II, 2° Policlinico Federico II
Napli, Italy, 80131
S. C. Farmacia
Napoli, Italy, 80131
Italy, Lombardia
Departimento Oncologia Medica
Milan, Lombardia, Italy, 20132
Unita Operativa di Oculistica: UO Oncologia Oculare e Immunopatologia oculare
Milan, Lombardia, Italy, 20132
Unita Operativa Radiologia
Milan, Lombardia, Italy, 20132
UO Anatomia e lstologia Patologica
Milan, Lombardia, Italy, 20132
UO Dermatologia e Cosmetologia
Milan, Lombardia, Italy, 20132
UO Medicina Nucleare
Milan, Lombardia, Italy, 20132
UOC Farmacia
Milan, Lombardia, Italy, 20132
Italy, TO
Dermatologia Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino
Torino, TO, Italy, 10126
Italy, Torino
S.S.D. Oncologia Polmonare
Orbassano, Torino, Italy, 10043
SCDO Cardiologia
Orbassano, Torino, Italy, 10043
SCDU Anatomia Patologica
Orbassano, Torino, Italy, 10043
SCDU Oftalmologia
Orbassano, Torino, Italy, 10043
SCDU Radiodiagnostica e S.S. Medicina Nucleare
Orbassano, Torino, Italy, 10043
Korea, Republic of
Samsung Medical Center - PPDS
Seoul, Korea, Republic of, 06351
Samsung Medical Center
Seoul, Korea, Republic of, 06351
Netherlands
Het Nederlands Kanker Instituut Antoni Van Leeuwenhoek Ziekenhuis
Amsterdam, Netherlands, 1066 CX
OLVG, locatie Oost; Ophthalmology department
Amsterdam, Netherlands, 1091 AC
Ophthalmology department
Groningen, Netherlands, 9713 GZ
University Medical Center Groningen
Groningen, Netherlands, 9713 GZ
Netherlands, Noord-holland
Het Nederlands Kanker Instituut Antoni Van Leeuwenhoek Ziekenhuis
Amsterdam, Noord-holland, Netherlands, 1066 CX
Spain
Hospital Clinic de Barcelona
Badalona, Spain, 08036
Hospital del Mar
Barcelona, Spain, 08003
CETIR Centro Medico Teknon
Barcelona, Spain, 08022
Hospital Quiron Salud Barcelona
Barcelona, Spain, 08023
Hospital Universitario Vall d'Hebron
Barcelona, Spain, 08035
Grupo Cardiologico Corpal (Hospital de la Cruz Roja)
Cordoba, Spain, 14004
Hospital Universitario Reina Sofia
Cordoba, Spain, 14004
Cetir
Esplugues de Llobregat, Spain, 08950
lnstitut Catala d'Oncologia_L'Hospitalet
L'Hospitalet, Spain, 08907
Hospital Universitario Ramon y Cajal
Madrid, Spain, 28034
Hospital Universitario 12 de Octubre
Madrid, Spain, 28041
Centro Hospitalario Integral Privado (CHIP)
Malaga, Spain, 29010
CERCO
Sevilla, Spain, 41009
Hospital Universitario Virgen Macarena
Sevilla, Spain, 41009
Hospital Universitario Virgen Del Rocio
Sevilla, Spain, 41013
Spain, Madrid
Hospital Universitario Puerta de Hierro - Majadahonda
Majadahonda, Madrid, Spain, 28222
Spain, Málaga
Hospital Regional Universitario de Malaga - Hospital General
Malaga, Málaga, Spain, 29010
Open or close this module IPDSharing
Plan to Share IPD: Yes
Pfizer will provide access to individual de-identified participant data and related study documents (e.g. protocol, Statistical Analysis Plan (SAP), Clinical Study Report (CSR)) upon request from qualified researchers, and subject to certain criteria, conditions, and exceptions. Further details on Pfizer's data sharing criteria and process for requesting access can be found at: https://www.pfizer.com/science/clinical_trials/trial_data_and_results/data_requests.
Supporting Information:
Time Frame:
Access Criteria:
URL: https://www.pfizer.com/science/clinical_trials/trial_data_and_results/data_requests
Open or close this module References
Links: Description: To obtain contact information for a study center near you, click here.
Available IPD/Information:
Open or close this module Document Section
Study Protocol
Document Date: September 24, 2021
Uploaded: 09/18/2023 15:00
File Name: Prot_000.pdf
Statistical Analysis Plan
Document Date: May 26, 2022
Uploaded: 09/18/2023 15:00
File Name: SAP_001.pdf
Study Results
Open or close this module Participant Flow
Recruitment Details
Pre-assignment Details
 
Arm/Group Title Treatment-Naive Previously Treated
Arm/Group Description Treatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal. Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.
Period Title: Overall Study
Started 59 39
Completed 0 0
Not Completed 59 39
Reason Not Completed
As of the PCD data-cut, 33 participants remained on-treatment. 25 8
Adverse Event 12 6
Disease progression (radiological) 17 15
Disease progression (clinical) 4 1
Patient decision 0 4
Investigator decision 0 1
Consent withdrawn 1 1
Death 0 3
Open or close this module Baseline Characteristics
Arm/Group TitleTreatment-NaivePreviously TreatedTotal
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Total of all reporting groups
Overall Number of Baseline Participants 59 39 98
Baseline Analysis Population Description
Age, Continuous
Median (Full Range)
Unit of measure: Years
Number Analyzed59 Participants39 Participants98 Participants
68(47 to 83)71(53 to 86)69.5(47 to 86)
Sex: Female, Male
Measure Type: Count of Participants
Unit of measure: Participants
Number Analyzed59 Participants39 Participants98 Participants
Female
33
55.93%
19
48.72%
52
53.06%
Male
26
44.07%
20
51.28%
46
46.94%
Race (NIH/OMB)
Measure Type: Count of Participants
Unit of measure: Participants
Number Analyzed59 Participants39 Participants98 Participants
American Indian or Alaska Native
1
1.69%
0
0%
1
1.02%
Asian
3
5.08%
4
10.26%
7
7.14%
Native Hawaiian or Other Pacific Islander
0
0%
0
0%
0
0%
Black or African American
1
1.69%
2
5.13%
3
3.06%
White
53
89.83%
33
84.62%
86
87.76%
More than one race
0
0%
0
0%
0
0%
Unknown or Not Reported
1
1.69%
0
0%
1
1.02%
Eastern Cooperation Oncology Group (ECOG) Performance Status [1]
Measure Type: Count of Participants
Unit of measure: Participants
Number Analyzed59 Participants39 Participants98 Participants
Grade 0
19
32.2%
7
17.95%
26
26.53%
Grade 1
40
67.8%
32
82.05%
72
73.47%
 
[1]Measure Description: ECOG Performance Status Scoring: Grade 0: Fully active, able to carry on all predisease performance without restriction; Grade 1: restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature; Grade 2: ambulatory and capable of all self-care but unable to carry out any work activities; up and about more than 50% of waking hours; Grade 3: capable of only limited self-care; confined to bed or chair more than 50% of waking hours; Grade 4: Completely disabled; cannot carry on any self-care; totally confined to bed or chair; Grade 5: dead.
Smoking Status [1]
Measure Type: Count of Participants
Unit of measure: Participants
Number Analyzed59 Participants39 Participants98 Participants
Current Smoking
8
13.56%
5
12.82%
13
13.27%
Former Smoking
33
55.93%
23
58.97%
56
57.14%
Never Smoking
18
30.51%
11
28.21%
29
29.59%
 
[1]Measure Description: Smoking history was collected at Screening by the Investigator or qualified designee.
Open or close this module Outcome Measures
1. Primary Outcome:
Title Percentage of Participants With Confirmed Objective Response (OR) as Determined by Independent Radiology Review (IRR)
Description Objective Response Rate (ORR) was defined as the percentage of participants who had achieved a confirmed best overall response (Complete Response [CR] or Partial Response [PR]) per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. Both CR and PR must be confirmed by repeat assessments performed no less than 4 weeks after the criteria for response were first met. CR was defined by the disappearance of all non-lymph node target lesions (where all target lesions were recorded with a length of 0 mm, and any pathological lymph nodes [recorded as target lesion] must have reduction in short axis to <10 mm) and complete disappearance of all non-target lesions (where all non-target lesions were marked "Absent", and all lymph nodes must be non-pathological in size [<10 mm in short axis]). PR was defined by a 30% or more decrease in sum of diameters (SOD) of target lesions, taking as reference the baseline SOD. ORR was calculated with the exact 2-sided Clopper-Pearson 95% CI.
Time Frame From date of the first dose of study intervention until documented progressive disease (PD) or start of new anticancer therapy (up to 36 months)
Outcome Measure Data
Analysis Population Description
Analysis population included all participants who received at least 1 dose of study intervention.
 
Arm/Group TitleTreatment-NaivePreviously Treated
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.
Overall Number of Participants Analyzed59 39
Number (95% Confidence Interval)
Unit of Measure: Percentage of Participants
74.6(61.6 to 85.0) 46.2(30.1 to 62.8)
2. Secondary Outcome:
Title Percentage of Participants With Confirmed Objective Response (OR) by Investigator Assessment
Description ORR was defined as the percentage of participants who had achieved a confirmed best overall response (CR or PR) per RECIST version 1.1. Both CR and PR must be confirmed by repeat assessments performed no less than 4 weeks after the criteria for response were first met. CR was defined by the disappearance of all non-lymph node target lesions (where all target lesions were recorded with a length of 0 mm, and any pathological lymph nodes [recorded as target lesion] must have reduction in short axis to <10 mm) and the complete disappearance of all non-target lesions (where all non-target lesions were marked "Absent", and all lymph nodes must be non-pathological in size [<10 mm in short axis]). PR was defined by a 30% or more decrease in SOD of target lesions, taking as reference the baseline SOD. ORR according to derived investigator assessment was calculated with the exact 2-sided Clopper-Pearson 95% CI.
Time Frame From date of the first dose of study intervention until documented PD or start of new anticancer therapy (up to 36 months)
Outcome Measure Data
Analysis Population Description
Analysis population included all participants who received at least 1 dose of study intervention.
 
Arm/Group TitleTreatment-NaivePreviously Treated
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.
Overall Number of Participants Analyzed59 39
Number (95% Confidence Interval)
Unit of Measure: Percentage of Participants
62.7(49.1 to 75.0) 41.0(25.6 to 57.9)
3. Secondary Outcome:
Title Duration of Response (DoR) by IRR and Investigator Assessments
Description DoR, based on IRR and Investigator assessments was defined as the time from the date of the first documented confirmed response (CR or PR) (by IRR and by Investigator, respectively) to the earliest date of disease progression, as determined by Investigator review of radiographic disease assessments and IRR per RECIST version 1.1, or death due to any cause. If a participant with a CR or PR had neither progressed nor died at the time of the analysis cutoff or at the start of any new anticancer therapy, the participant was censored at the date of last adequate tumor assessment. CR was defined by the disappearance of all non-lymph node target lesions and complete disappearance of all non-target lesions. PR was defined by a 30% or more decrease in SOD of target lesions, taking as reference the baseline SOD. DoR was calculated for participants who had achieved a confirmed overall response (CR or PR). The estimate of the DoR survival function was constructed using the Kaplan-Meier method.
Time Frame From the date of the first confirmed documented response (CR or PR) to the earliest date of disease progression or death due to any cause (up to 36 months)
Anticipated Reporting Date June 2025
Outcome Measure Data Not Reported
4. Secondary Outcome:
Title Disease Control Rate (DCR) by IRR and Investigator Assessments
Description DCR was defined as the percentage of participants who had achieved a confirmed overall response of CR, PR or stable disease (SD), as determined by Investigator review of radiographic disease assessments and IRR per RECIST version 1.1 after 24 weeks (≥168 days) from the date of first dose of study intervention. CR was defined by the disappearance of all non-lymph node target lesions and complete disappearance of all non-target lesions. PR was defined by a 30% or more decrease in SOD of target lesions, taking as reference the baseline SOD. SD was assigned when neither sufficient shrinkage to qualify for CR or PR, nor sufficient increase to qualify for PD was observed, taking as reference the nadir. DCR was calculated along with the exact 2-sided Clopper-Pearson 95% CI.
Time Frame After 24 Weeks (≥168 days) from the date of first dose of study intervention
Outcome Measure Data
Analysis Population Description
Analysis population included all participants who received at least 1 dose of study intervention.
 
Arm/Group TitleTreatment-NaivePreviously Treated
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.
Overall Number of Participants Analyzed59 39
Number (95% Confidence Interval)
Unit of Measure: Percentage of Participants
DCR After 24 Weeks by IRR
64.4(50.9 to 76.4) 41.0(25.6 to 57.9)
DCR After 24 Weeks by Investigator Assessment
67.8(54.4 to 79.4) 48.7(32.4 to 65.2)
5. Secondary Outcome:
Title Progression-free Survival (PFS) by IRR and Investigator Assessments
Description PFS was defined as the time from the date of first dose of study intervention to the earliest date of disease progression, as determined by IRR and Investigator review of radiographic disease assessments per RECIST version 1.1, or death due to any cause, whichever occurred first. PD was defined by a 20% or more increase in the SOD of target lesions relative to nadir (smallest SOD considering baseline and all assessments prior to the time point under evaluation), with a minimum absolute increase of 5 mm relative to nadir; PD was assigned if any non-target lesion was marked "unequivocal progression". If a participant had not had a PFS event at the time of the analysis cutoff or at the start of any new anticancer therapy, PFS was censored at the date of last adequate tumor assessment. PFS (months) = [(date of event or censoring - date of first dose) +1]/30.4375. The survival distribution function for PFS was estimated using the Kaplan-Meier method.
Time Frame From the date of first dose of study drug to the earliest date of disease progression, or death due to any cause (up to 36 months)
Anticipated Reporting Date June 2025
Outcome Measure Data Not Reported
6. Secondary Outcome:
Title Time to Response (TTR) by IRR and Investigator Assessments
Description TTR based on IRR and Investigator assessments was defined, for participants with an objective response, as the time, in months, from the date of first dose to the first documentation of objective response (CR or PR) which was subsequently confirmed (by IRR and by Investigator, respectively). TTR was calculated for the subgroup of participants with a confirmed objective tumor response.
Time Frame From the date of first dose to the first documentation of confirmed objective response (CR or PR) (up to 36 months)
Outcome Measure Data
Analysis Population Description
Analysis population included all participants who received at least 1 dose of study intervention and who had confirmed objective tumor response (CR or PR). Number of participants analyzed signifies number of participants who were evaluable for this outcome measure. Number analyzed refers to number of participants evaluable for specified rows of categories.
   
Arm/Group TitleTreatment-NaivePreviously Treated
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.
Overall Number of Participants Analyzed59 39
Median (Full Range)
Unit of Measure: Months
 
TTR by IRR
Number Analyzed Participants Participants
1.86(1.1 to 19.1) 1.74(1.2 to 7.3)
TTR by Investigator Assessment
Number Analyzed Participants Participants
1.84(1.4 to 14.0) 1.81(0.8 to 9.2)
7. Secondary Outcome:
Title Kaplan-Meier Estimates of Overall Survival (OS)
Description OS was defined as the time from the date of first dose of study intervention to the date of death due to any cause. If a death had not been observed by the date of the analysis cutoff, OS was censored at the date of last contact. The survival distribution function for OS was estimated using the Kaplan-Meier method.
Time Frame The time from the date of first dose of study intervention to the date of death due to any cause (up to 36 months)
Anticipated Reporting Date June 2025
Outcome Measure Data Not Reported
8. Secondary Outcome:
Title Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
Description TEAE was a treatment-emergent adverse event that occurred during the on-treatment period. The on-treatment period was defined as the time from the first dose date of study intervention to the last dose of study drug administration date (when both drugs were permanently discontinued) +30 days or the earliest date of subsequent anti-cancer drug therapy minus 1 day, whichever occurred first. Relatedness to study intervention was determined by the investigator. The investigator made an assessment of intensity for each AE reported during the study according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.03: Grade 3 events = severe AEs; Grade 4 events = life-threatening consequences, urgent intervention indicated; Grade 5 events = death related to AEs.
Time Frame From the time the participant provided informed consent, through and including a minimum of 30 calendar days, after the last administration of the study intervention, and the safety follow-up visit (30 days [±7 days] after the EOT visit) (up to 36 months)
Outcome Measure Data
Analysis Population Description
Safety Analysis Set included all participants who received at least 1 dose of study intervention.
 
Arm/Group TitleTreatment-NaivePreviously TreatedTotal
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Sum of all participants in the Study C4221008 (ARRAY-818-202)
Overall Number of Participants Analyzed59 39 98
Measure Type: Count of Participants
Unit of Measure: Participants
Participants With TEAEs
59
100%
38
97.4%
97
99%
Participants With Treatment-related TEAEs
58
98.3%
34
87.2%
92
93.9%
Participants with Maximum Grade 3 or 4 TEAEs
37
62.7%
18
46.2%
55
56.1%
Participants with Maximum Treatment-related Grade 3 or 4 TEAEs
27
45.8%
13
33.3%
40
40.8%
Participants With Grade 5 TEAEs
5
8.5%
7
17.9%
12
12.2%
Participants With Treatment-related Grade 5 TEAEs
1
1.7%
0
0%
1
1%
9. Secondary Outcome:
Title Number of Participants With Shifts From Grade ≤2 at Baseline to Grade 3 or 4 at Post-baseline in Hematology Laboratory Test Values Based on CTCAE Grade
Description Blood and urine samples for the laboratory tests. A central laboratory will perform all clinical laboratory assessments. Baseline was the last available assessment performed prior to the study intervention start date/time.
Time Frame Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Day 1 ±3 Days in Subsequent Cycles, End of Treatment (EOT) ±3 Days and Safety Follow-up Visit (30 Days [±7 Days] After the EOT Visit) (Up to 36 Months)
Outcome Measure Data
Analysis Population Description
Safety Analysis Set included all participants who received at least 1 dose of study intervention. Number of participants analyzed signifies number of participants who were evaluable for this outcome measure. Number analyzed refers to number of participants evaluable for specified rows of categories.
   
Arm/Group TitleTreatment-NaivePreviously TreatedTotal
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Sum of all participants in the Study C4221008 (ARRAY-818-202)
Overall Number of Participants Analyzed59 39 98
Measure Type: Count of Participants
Unit of Measure: Participants
 
Anemia - Grade 3
Number Analyzed Participants Participants Participants
9
15.3%
1
2.6%
10
10.2%
Anemia - Grade 4
Number Analyzed Participants Participants Participants
NA [1] NA [1] NA [1]
Hemoglobin increased - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hemoglobin increased - Grade 4
Number Analyzed Participants Participants Participants
NA [1] NA [1] NA [1]
Leukocytosis - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Leukocytosis - Grade 4
Number Analyzed Participants Participants Participants
NA [1] NA [1] NA [1]
Lymphocyte count decreased - Grade 3
Number Analyzed Participants Participants Participants
2
3.4%
3
7.7%
5
5.1%
Lymphocyte count decreased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Lymphocyte count increased - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Lymphocyte count increased - Grade 4
Number Analyzed Participants Participants Participants
NA [1] NA [1] NA [1]
Neutrophil count decreased - Grade 3
Number Analyzed Participants Participants Participants
1
1.7%
0
0%
1
1%
Neutrophil count decreased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Platelet count decreased - Grade 3
Number Analyzed Participants Participants Participants
0
0%
1
2.6%
1
1%
Platelet count decreased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
White blood cell decreased - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
White blood cell decreased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
[1]NA Explanation: CTCAE Grade 4 was not defined for this parameter.

Quality Control Review Comment provided by the National Library of Medicine:

  1. The explanation provided is not sufficient to understand why one or more values are not available.
10. Secondary Outcome:
Title Number of Participants With Shifts From Grade ≤2 at Baseline to Grade 3 or 4 at Post-baseline in Chemistry Laboratory Test Values Based on CTCAE Grade
Description Blood and urine samples for the laboratory tests. A central laboratory will perform all clinical laboratory assessments. Baseline was the last available assessment performed prior to the study intervention start date/time.
Time Frame Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Day 1 ±3 Days in Subsequent Cycles, EOT ±3 Days and Safety Follow-up Visit (30 Days [±7 Days] After the EOT Visit) (Up to 36 Months)
Outcome Measure Data
Analysis Population Description
Safety Analysis Set included all participants who received at least 1 dose of study intervention. Number of participants analyzed signifies number of participants who were evaluable for this outcome measure. Number analyzed refers to number of participants evaluable for specified rows of categories.
   
Arm/Group TitleTreatment-NaivePreviously TreatedTotal
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Sum of all participants in the Study C4221008 (ARRAY-818-202)
Overall Number of Participants Analyzed59 39 98
Measure Type: Count of Participants
Unit of Measure: Participants
 
Alanine aminotransferase increased - Grade 3
Number Analyzed Participants Participants Participants
6
10.2%
2
5.1%
8
8.2%
Alanine aminotransferase increased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Alkaline phosphatase increased - Grade 3
Number Analyzed Participants Participants Participants
2
3.4%
1
2.6%
3
3.1%
Alkaline phosphatase increased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Aspartate aminotransferase increased - Grade 3
Number Analyzed Participants Participants Participants
7
11.9%
2
5.1%
9
9.2%
Aspartate aminotransferase increased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Blood bilirubin increased - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Blood bilirubin increased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Creatine kinase increased - Grade 3
Number Analyzed Participants Participants Participants
2
3.4%
1
2.6%
3
3.1%
Creatine kinase increased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Creatinine increased - Grade 3
Number Analyzed Participants Participants Participants
3
5.1%
0
0%
3
3.1%
Creatinine increased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypercalcemia - Grade 3
Number Analyzed Participants Participants Participants
1
1.7%
1
2.6%
2
2%
Hypercalcemia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hyperglycemia - Grade 3
Number Analyzed Participants Participants Participants
5
8.5%
1
2.6%
6
6.1%
Hyperglycemia - Grade 4
Number Analyzed Participants Participants Participants
NA [1] NA [1] NA [1]
Hyperkalemia - Grade 3
Number Analyzed Participants Participants Participants
2
3.4%
0
0%
2
2%
Hyperkalemia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypermagnesemia - Grade 3
Number Analyzed Participants Participants Participants
0
0%
1
2.6%
1
1%
Hypermagnesemia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypernatremia - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypernatremia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypoalbuminemia - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypoalbuminemia - Grade 4
Number Analyzed Participants Participants Participants
NA [1] NA [1] NA [1]
Hypocalcemia - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypocalcemia - Grade 4
Number Analyzed Participants Participants Participants
2
3.4%
0
0%
2
2%
Hypoglycemia - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypoglycemia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypokalemia - Grade 3
Number Analyzed Participants Participants Participants
0
0%
1
2.6%
1
1%
Hypokalemia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypomagnesemia - Grade 3
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypomagnesemia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hyponatremia - Grade 3
Number Analyzed Participants Participants Participants
7
11.9%
2
5.1%
9
9.2%
Hyponatremia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Hypophosphatemia - Grade 3
Number Analyzed Participants Participants Participants
1
1.7%
0
0%
1
1%
Hypophosphatemia - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
Lipase increased - Grade 3
Number Analyzed Participants Participants Participants
8
13.6%
3
7.7%
11
11.2%
Lipase increased - Grade 4
Number Analyzed Participants Participants Participants
2
3.4%
0
0%
2
2%
Serum amylase increased - Grade 3
Number Analyzed Participants Participants Participants
1
1.7%
0
0%
1
1%
Serum amylase increased - Grade 4
Number Analyzed Participants Participants Participants
0
0%
0
0%
0
0%
[1]NA Explanation: CTCAE Grade 4 was not defined for this parameter.

Quality Control Review Comment provided by the National Library of Medicine:

  1. The explanation provided is not sufficient to understand why one or more values are not available.
11. Secondary Outcome:
Title Number of Participants With Notable Abnormal Vital Signs
Description

Vital sign measurements were taken before blood collection for laboratory tests or at least 30 minutes after blood collection for laboratory tests, and were measured per institutional standards. Vital sign assessments included temperature, pulse rate, respiratory rate, and blood pressure (assessed in a recumbent, semi recumbent, or sitting position).

The criteria of notably abnormal vital signs are listed below:

Systolic blood pressure (mmHg): high: ≥160 mmHg with increase from baseline of ≥20 mmHg; low: ≤90 mmHg with decrease from baseline of ≥20 mmHg.

Diastolic blood pressure (mmHg): high: ≥100 mmHg with increase from baseline of ≥15 mmHg; low: ≤50 mmHg with decrease from baseline of ≥15 mmHg.

Pulse rate (bpm): high: ≥120 bpm with increase from baseline of ≥15 bpm; low: ≤50 bpm with decrease from baseline of ≥15 bpm.

Weight (kg): high: ≥10% increase from baseline; low: ≥20% decrease from baseline.

Temperature (°C): high: ≥37.5 °C; low: ≤36 °C.

Time Frame Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Day 1 ±3 Days of Subsequent Cycles, EOT ±3 Days, and Safety Follow-up Visit (30 Days [±7 Days] After the EOT Visit) (Up to 36 Months)
Outcome Measure Data
Analysis Population Description
Analysis population included all participants who received at least 1 dose of study intervention and had baseline and post-baseline vital sign values. Number of participants analyzed signifies number of participants who were evaluable for this outcome measure. Number analyzed refers to number of participants evaluable for specified rows of categories.
   
Arm/Group TitleTreatment-NaivePreviously TreatedTotal
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Sum of all participants in the Study C4221008 (ARRAY-818-202)
Overall Number of Participants Analyzed59 39 98
Measure Type: Count of Participants
Unit of Measure: Participants
 
Systolic blood pressure - high
Number Analyzed Participants Participants Participants
10
16.9%
4
10.3%
14
14.3%
Systolic blood pressure - low
Number Analyzed Participants Participants Participants
5
8.5%
2
5.1%
7
7.1%
Diastolic blood pressure - high
Number Analyzed Participants Participants Participants
5
8.5%
1
2.6%
6
6.1%
Diastolic blood pressure - low
Number Analyzed Participants Participants Participants
2
3.4%
2
5.1%
4
4.1%
Pulse rate - high
Number Analyzed Participants Participants Participants
2
3.4%
6
15.4%
8
8.2%
Pulse rate - low
Number Analyzed Participants Participants Participants
1
1.7%
1
2.6%
2
2%
Weight - high
Number Analyzed Participants Participants Participants
17
28.8%
4
10.3%
21
21.4%
Weight - low
Number Analyzed Participants Participants Participants
1
1.7%
0
0%
1
1%
Temperature - high
Number Analyzed Participants Participants Participants
8
13.6%
3
7.7%
11
11.2%
Temperature - low
Number Analyzed Participants Participants Participants
25
42.4%
10
25.6%
35
35.7%
12. Secondary Outcome:
Title Number of Participants With Notable ECG (QTcF) Values
Description The QTcF increase from baseline >30/60 msec and new QTcF >450/480/500 msec were defined as clinically notable ECG criteria. Triplicate ECG measurements were obtained. For new abnormal post-baseline values, the table below presents the number of participants with both non-missing baseline and post-baseline values, and baseline values not meeting the criteria. For abnormal changes from baseline, the table below presents the number of participants with both non-missing baseline and post-baseline evaluations. Baseline was defined as the average of the machine-read triplicate ECG measurements taken pre-dose on Day 1. Change from baseline was post-baseline - baseline values.
Time Frame Screening, Cycle 1 Day 1, Cycle 2 Day 1 ±3 Days, Every 12 Weeks ±7 Days, and EOT ±3 Days (Up to 36 Months)
Outcome Measure Data
Analysis Population Description
Safety Analysis Set included all participants who received at least 1 dose of study intervention. Number of participants analyzed signifies number of participants who were evaluable for this outcome measure. Number analyzed refers to number of participants evaluable for specified rows of categories.
   
Arm/Group TitleTreatment-NaivePreviously TreatedTotal
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Sum of all participants in the Study C4221008 (ARRAY-818-202)
Overall Number of Participants Analyzed59 39 98
Measure Type: Count of Participants
Unit of Measure: Participants
 
New QTcF >450 msec
Number Analyzed Participants Participants Participants
12
20.3%
7
17.9%
19
19.4%
New QTcF >480 msec
Number Analyzed Participants Participants Participants
5
8.5%
5
12.8%
10
10.2%
New QTcF >500 msec
Number Analyzed Participants Participants Participants
1
1.7%
1
2.6%
2
2%
QTcF Increase from baseline >30 msec
Number Analyzed Participants Participants Participants
20
33.9%
9
23.1%
29
29.6%
QTcF Increase from baseline >60 msec
Number Analyzed Participants Participants Participants
3
5.1%
4
10.3%
7
7.1%
13. Secondary Outcome:
Title Number of Participants With the Worst Post-baseline Left Ventricular Ejection Fraction (LVEF) Values Based on CTCAE Grade
Description

Cardiac ejection fraction was assessed by transthoracic echocardiogram (ECHO) or multigated acquisition (MUGA). Participants who developed signs/symptoms of congestive heart failure (CHF) at any point during the study were required to have an evaluation of LVEF measurements by ECHO or MUGA and were monitored per institutional guidelines.

Participants were considered as having a LVEF abnormality if the worst post-value was CTCAE Grade 2, 3 or 4 according to the following classification:

Grade 0: Non-missing value below Grade 2. Grade 2: LVEF between 40% and 50%, inclusive, or absolute change from baseline between -10% and < -20%.

Grade 3: LVEF between 20% and 39%, inclusive, or absolute change from baseline ≤ -20%.

Grade 4: LVEF lower than 20%. Baseline was defined as the last available and valid assessment before or on the start date of study intervention.

Time Frame Screening, Cycle 2 Day 1 ±3 Days, Every 12 Weeks ±7 Days, and EOT ±3 Days (Up to 36 Months)
Outcome Measure Data
Analysis Population Description
Analysis Population included all participants who received at least 1 dose of study intervention and had both baseline and post-baseline LVEF values. Number of participants analyzed signifies number of participants who were evaluable for this outcome measure. Number analyzed refers to number of participants evaluable for specified rows of categories.
 
Arm/Group TitleTreatment-NaivePreviously TreatedTotal
Arm/Group DescriptionTreatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal.Sum of all participants in the Study C4221008 (ARRAY-818-202)
Overall Number of Participants Analyzed56 33 89
Measure Type: Count of Participants
Unit of Measure: Participants
Grade 0
45
80.4%
24
72.7%
69
77.5%
Grade 2
11
19.6%
7
21.2%
18
20.2%
Grade 3
0
0%
2
6.1%
2
2.2%
Grade 4
0
0%
0
0%
0
0%
Open or close this module Adverse Events
 
Time Frame From the time the participant provided informed consent, through and including a minimum of 30 calendar days, after the last administration of the study intervention, and the safety follow-up visit (30 days [±7 days] after the EOT visit) (up to 36 months)
Adverse Event Reporting Description Same event may appear as both an AE and a serious AE (SAE). However, what is presented are distinct events. An event may be categorized as serious in 1 participant and as non-serious in another participant, or 1 participant may have experienced both serious and non-serious event during the study. Safety analysis set included all participants who received at least 1 dose of study intervention.
 
Arm/Group Title Treatment-Naive Previously Treated Total
Arm/Group Description Treatment-naïve participants with BRAF V600E-mutant metastatic NSCLC were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal. Patients who had received 1) first-line treatment with standard platinum-based chemotherapy, or 2) first-line treatment with an anti-PD-1/PD-L1 inhibitor given alone, or in combination with platinum-based chemotherapy, or in combination with immunotherapy (eg, ipilimumab) with or without platinum-based chemotherapy were enrolled to receive encorafenib 450 mg QD and binimetinib 45 mg BID was to be administered in 28-day (±3 days) cycles and continued until the participants met the protocol-defined criteria for treatment withdrawal. Sum of all participants in the Study C4221008 (ARRAY-818-202)
All-Cause Mortality
  Treatment-NaivePreviously TreatedTotal
 Affected / At Risk (%)Affected / At Risk (%)Affected / At Risk (%)
Total 5 / 59 (8.47%)7 / 39 (17.95%)12 / 98 (12.24%)
Serious Adverse Events
  Treatment-NaivePreviously TreatedTotal
 Affected / At Risk (%)Affected / At Risk (%)Affected / At Risk (%)
Total 25 / 59 (42.37%)17 / 39 (43.59%)42 / 98 (42.86%)
Blood and lymphatic system disorders
Anaemia ∗ A 3 / 59 (5.08%)0 / 39 (0%)3 / 98 (3.06%)
Disseminated intravascular coagulation ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Cardiac disorders
Atrial fibrillation ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Myocardial infarction ∗ A 1 / 59 (1.69%)1 / 39 (2.56%)2 / 98 (2.04%)
Pericardial effusion ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Supraventricular tachyarrhythmia ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Tachycardia ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Eye disorders
Retinal detachment ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Gastrointestinal disorders
Colitis ∗ A 4 / 59 (6.78%)0 / 39 (0%)4 / 98 (4.08%)
Constipation ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Diarrhoea ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Duodenal ulcer ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Gastrointestinal haemorrhage ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Nausea ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Small intestinal haemorrhage ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Upper gastrointestinal haemorrhage ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
General disorders
Asthenia ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Disease progression ∗ A 2 / 59 (3.39%)4 / 39 (10.26%)6 / 98 (6.12%)
Oedema peripheral ∗ A 2 / 59 (3.39%)0 / 39 (0%)2 / 98 (2.04%)
Hepatobiliary disorders
Cholecystitis ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Cholelithiasis ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Infections and infestations
COVID-19 ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
COVID-19 pneumonia ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Cellulitis ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Clostridium difficile colitis ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Device related infection ∗ A 1 / 59 (1.69%)1 / 39 (2.56%)2 / 98 (2.04%)
Empyema ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Herpes zoster ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Ophthalmic herpes zoster ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Pneumonia ∗ A 2 / 59 (3.39%)0 / 39 (0%)2 / 98 (2.04%)
Pneumonia mycoplasmal ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Respiratory tract infection ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Urinary tract infection ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Urosepsis ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Injury, poisoning and procedural complications
Femur fracture ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Head injury ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Hip fracture ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Upper limb fracture ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Investigations
Blood creatinine increased ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Metabolism and nutrition disorders
Hyponatraemia ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Musculoskeletal and connective tissue disorders
Back pain ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Neoplasm progression ∗ A 2 / 59 (3.39%)2 / 39 (5.13%)4 / 98 (4.08%)
Squamous cell carcinoma of skin ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Nervous system disorders
Cerebrovascular accident ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Cognitive disorder ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Haemorrhage intracranial ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Hemiparesis ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Presyncope ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Seizure ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Psychiatric disorders
Confusional state ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Hallucination, auditory ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Renal and urinary disorders
Acute kidney injury ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Respiratory, thoracic and mediastinal disorders
Chronic respiratory failure ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Dyspnoea ∗ A 0 / 59 (0%)3 / 39 (7.69%)3 / 98 (3.06%)
Haemoptysis ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Haemothorax ∗ A 2 / 59 (3.39%)0 / 39 (0%)2 / 98 (2.04%)
Pleural effusion ∗ A 1 / 59 (1.69%)1 / 39 (2.56%)2 / 98 (2.04%)
Pneumothorax ∗ A 0 / 59 (0%)1 / 39 (2.56%)1 / 98 (1.02%)
Vascular disorders
Hypertension ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Peripheral embolism ∗ A 1 / 59 (1.69%)0 / 39 (0%)1 / 98 (1.02%)
Indicates events were collected by non-systematic methods.
ATerm from vocabulary, MedDRA version 25.0
Other (Not Including Serious) Adverse Events
Frequency Threshold for Reporting Other Adverse Events 5%
  Treatment-NaivePreviously TreatedTotal
 Affected / At Risk (%)Affected / At Risk (%)Affected / At Risk (%)
Total 59 / 59 (100%)38 / 39 (97.44%)97 / 98 (98.98%)
Blood and lymphatic system disorders
Anaemia ∗ A 20 / 59 (33.9%)10 / 39 (25.64%)30 / 98 (30.61%)
Eye disorders
Vision blurred ∗ A 14 / 59 (23.73%)6 / 39 (15.38%)20 / 98 (20.41%)
Gastrointestinal disorders
Abdominal pain ∗ A 14 / 59 (23.73%)6 / 39 (15.38%)20 / 98 (20.41%)
Abdominal pain upper ∗ A 4 / 59 (6.78%)4 / 39 (10.26%)8 / 98 (8.16%)
Constipation ∗ A 18 / 59 (30.51%)8 / 39 (20.51%)26 / 98 (26.53%)
Diarrhoea ∗ A 29 / 59 (49.15%)20 / 39 (51.28%)49 / 98 (50%)
Nausea ∗ A 37 / 59 (62.71%)19 / 39 (48.72%)56 / 98 (57.14%)
Vomiting ∗ A 23 / 59 (38.98%)13 / 39 (33.33%)36 / 98 (36.73%)
General disorders
Asthenia ∗ A 7 / 59 (11.86%)9 / 39 (23.08%)16 / 98 (16.33%)
Chills ∗ A 3 / 59 (5.08%)3 / 39 (7.69%)6 / 98 (6.12%)
Fatigue ∗ A 29 / 59 (49.15%)16 / 39 (41.03%)45 / 98 (45.92%)
Malaise ∗ A 5 / 59 (8.47%)1 / 39 (2.56%)6 / 98 (6.12%)
Non-cardiac chest pain ∗ A 6 / 59 (10.17%)2 / 39 (5.13%)8 / 98 (8.16%)
Oedema peripheral ∗ A 12 / 59 (20.34%)8 / 39 (20.51%)20 / 98 (20.41%)
Pyrexia ∗ A 16 / 59 (27.12%)6 / 39 (15.38%)22 / 98 (22.45%)
Infections and infestations
COVID-19 ∗ A 5 / 59 (8.47%)2 / 39 (5.13%)7 / 98 (7.14%)
Investigations
Alanine aminotransferase increased ∗ A 10 / 59 (16.95%)3 / 39 (7.69%)13 / 98 (13.27%)
Amylase increased ∗ A 4 / 59 (6.78%)2 / 39 (5.13%)6 / 98 (6.12%)
Aspartate aminotransferase increased ∗ A 11 / 59 (18.64%)4 / 39 (10.26%)15 / 98 (15.31%)
Blood alkaline phosphatase increased ∗ A 9 / 59 (15.25%)2 / 39 (5.13%)11 / 98 (11.22%)
Blood creatine phosphokinase increased ∗ A 10 / 59 (16.95%)5 / 39 (12.82%)15 / 98 (15.31%)
Blood creatinine increased ∗ A 8 / 59 (13.56%)7 / 39 (17.95%)15 / 98 (15.31%)
Ejection fraction decreased ∗ A 4 / 59 (6.78%)4 / 39 (10.26%)8 / 98 (8.16%)
Lipase increased ∗ A 11 / 59 (18.64%)4 / 39 (10.26%)15 / 98 (15.31%)
Platelet count decreased ∗ A 2 / 59 (3.39%)4 / 39 (10.26%)6 / 98 (6.12%)
SARS-CoV-2 test positive ∗ A 4 / 59 (6.78%)4 / 39 (10.26%)8 / 98 (8.16%)
Weight decreased ∗ A 6 / 59 (10.17%)1 / 39 (2.56%)7 / 98 (7.14%)
Weight increased ∗ A 10 / 59 (16.95%)1 / 39 (2.56%)11 / 98 (11.22%)
Metabolism and nutrition disorders
Decreased appetite ∗ A 12 / 59 (20.34%)2 / 39 (5.13%)14 / 98 (14.29%)
Dehydration ∗ A 2 / 59 (3.39%)3 / 39 (7.69%)5 / 98 (5.1%)
Hyperglycaemia ∗ A 4 / 59 (6.78%)3 / 39 (7.69%)7 / 98 (7.14%)
Hyperkalaemia ∗ A 6 / 59 (10.17%)1 / 39 (2.56%)7 / 98 (7.14%)
Hypoalbuminaemia ∗ A 4 / 59 (6.78%)3 / 39 (7.69%)7 / 98 (7.14%)
Hyponatraemia ∗ A 7 / 59 (11.86%)5 / 39 (12.82%)12 / 98 (12.24%)
Musculoskeletal and connective tissue disorders
Arthralgia ∗ A 9 / 59 (15.25%)6 / 39 (15.38%)15 / 98 (15.31%)
Back pain ∗ A 11 / 59 (18.64%)8 / 39 (20.51%)19 / 98 (19.39%)
Muscle spasms ∗ A 10 / 59 (16.95%)2 / 39 (5.13%)12 / 98 (12.24%)
Muscular weakness ∗ A 5 / 59 (8.47%)2 / 39 (5.13%)7 / 98 (7.14%)
Myalgia ∗ A 6 / 59 (10.17%)4 / 39 (10.26%)10 / 98 (10.2%)
Pain in extremity ∗ A 6 / 59 (10.17%)7 / 39 (17.95%)13 / 98 (13.27%)
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Seborrhoeic keratosis ∗ A 5 / 59 (8.47%)2 / 39 (5.13%)7 / 98 (7.14%)
Nervous system disorders
Dizziness ∗ A 10 / 59 (16.95%)6 / 39 (15.38%)16 / 98 (16.33%)
Dysgeusia ∗ A 5 / 59 (8.47%)3 / 39 (7.69%)8 / 98 (8.16%)
Headache ∗ A 6 / 59 (10.17%)4 / 39 (10.26%)10 / 98 (10.2%)
Paraesthesia ∗ A 5 / 59 (8.47%)0 / 39 (0%)5 / 98 (5.1%)
Psychiatric disorders
Insomnia ∗ A 7 / 59 (11.86%)3 / 39 (7.69%)10 / 98 (10.2%)
Renal and urinary disorders
Pollakiuria ∗ A 6 / 59 (10.17%)0 / 39 (0%)6 / 98 (6.12%)
Respiratory, thoracic and mediastinal disorders
Cough ∗ A 11 / 59 (18.64%)6 / 39 (15.38%)17 / 98 (17.35%)
Dyspnoea ∗ A 16 / 59 (27.12%)7 / 39 (17.95%)23 / 98 (23.47%)
Productive cough ∗ A 8 / 59 (13.56%)3 / 39 (7.69%)11 / 98 (11.22%)
Skin and subcutaneous tissue disorders
Alopecia ∗ A 9 / 59 (15.25%)3 / 39 (7.69%)12 / 98 (12.24%)
Dry skin ∗ A 11 / 59 (18.64%)2 / 39 (5.13%)13 / 98 (13.27%)
Hair texture abnormal ∗ A 5 / 59 (8.47%)1 / 39 (2.56%)6 / 98 (6.12%)
Hyperkeratosis ∗ A 4 / 59 (6.78%)1 / 39 (2.56%)5 / 98 (5.1%)
Pruritus ∗ A 7 / 59 (11.86%)8 / 39 (20.51%)15 / 98 (15.31%)
Rash ∗ A 6 / 59 (10.17%)5 / 39 (12.82%)11 / 98 (11.22%)
Rash maculo-papular ∗ A 5 / 59 (8.47%)3 / 39 (7.69%)8 / 98 (8.16%)
Vascular disorders
Hypertension ∗ A 8 / 59 (13.56%)2 / 39 (5.13%)10 / 98 (10.2%)
Indicates events were collected by non-systematic methods.
ATerm from vocabulary, MedDRA version 25.0
Open or close this module Limitations and Caveats
[Not specified]
Open or close this module More Information
Certain Agreements:
Principal Investigators are NOT employed by the organization sponsoring the study.

There IS an agreement between the Principal Investigator and the Sponsor (or its agents) that restricts the PI's rights to discuss or publish trial results after the trial is completed.

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/Official Title:
Pfizer ClinicalTrials.gov Call Center
Organization:
Pfizer Inc.
Phone:
1-800-718-1021
Email:
ClinicalTrials.gov_Inquiries@pfizer.com

Scroll up to access the controls Scroll to the Study top

U.S. National Library of Medicine | U.S. National Institutes of Health | U.S. Department of Health & Human Services