Study of Binimetinib + Nivolumab Plus or Minus Ipilimumab in Patients With Previously Treated Microsatellite-stable (MSS) Metastatic Colorectal Cancer With RAS Mutation

Sponsor
Pfizer (Industry)
Overall Status
Completed
CT.gov ID
NCT03271047
Collaborator
Bristol-Myers Squibb (Industry)
75
50
4
40.3
1.5
0

Study Details

Study Description

Brief Summary

This is a multicenter, open-label, Phase 1B/2 study to evaluate the safety and assess the preliminary anti-tumor activity of binimetinib administered in combination with nivolumab or nivolumab + ipilimumab in adult patients with advanced metastatic colorectal cancer (mCRC) with microsatellite stable (MSS) disease and presence of a RAS mutation that have received at least one prior line of therapy and no more than 2 prior lines of therapy. The study contains a Phase 1b period to determine the maximum tolerated dose (MTD) and recommended Phase 2 dose (RP2D) and schedule of binimetinib followed by a randomized Phase 2 period to assess the efficacy of the combinations.

Condition or Disease Intervention/Treatment Phase
Phase 2

Study Design

Study Type:
Interventional
Actual Enrollment :
75 participants
Allocation:
Non-Randomized
Intervention Model:
Parallel Assignment
Intervention Model Description:
In phase 1 it is sequential and then in phase 2 it is parallel.In phase 1 it is sequential and then in phase 2 it is parallel.
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
An Open-label Phase 1b/2 Study of Binimetinib Administered in Combination With Nivolumab or Nivolumab Plus Ipilimumab in Patients With Previously Treated Microsatellite-stable (MSS) Metastatic Colorectal Cancer With RAS Mutation
Actual Study Start Date :
Oct 18, 2017
Actual Primary Completion Date :
Oct 13, 2020
Actual Study Completion Date :
Feb 25, 2021

Arms and Interventions

Arm Intervention/Treatment
Experimental: Phase 1b / Arm 1A

binimetinib + nivolumab

Drug: binimetinib
Orally, twice daily.

Drug: nivolumab
Intravenously (IV) every 4 weeks (Q4W)

Experimental: Phase 1b / Arm 1B

binimetinib + nivolumab + ipilimumab

Drug: binimetinib
Orally, twice daily.

Drug: nivolumab
Intravenously (IV) every 4 weeks (Q4W)

Drug: ipilimumab
intravenously (IV) every 8 weeks (Q8W)

Experimental: Phase 2 / Arm 2A

binimetinib + nivolumab

Drug: binimetinib
Orally, twice daily.

Drug: nivolumab
Intravenously (IV) every 4 weeks (Q4W)

Experimental: Phase 2 / Arm 2B

binimetinib + nivolumab + ipilimumab

Drug: binimetinib
Orally, twice daily.

Drug: nivolumab
Intravenously (IV) every 4 weeks (Q4W)

Drug: ipilimumab
intravenously (IV) every 8 weeks (Q8W)

Outcome Measures

Primary Outcome Measures

  1. Phase 1b: Number of Participants With Dose-Limiting Toxicities (DLT) [Cycle 1: Day 1 up to Day 28]

    DLT:Adverse event(AE)/abnormal laboratory assessed unrelated-disease,disease progression,intercurrent illness/concomitant medication/therapies resulting inability tolerate 75% dose intensity in Cycle 1.Total bilirubin(TBL)grade(G)>=3 (>3.0*upper limit of normal[ULN)]);AST/ALT>5-8*ULN>5 days,>8*ULN,>3*ULN concurrent TBL>2*ULN;G>=3 serum creatinine,CK elevation,ECG QTcF prolonged,G3 troponin,electrolyte>72 hours,G3/4 amylase/lipase.G4 ANC,platelet count>7 days;G3/4 platelet count,other AE except lymphopenia.G>=3 retinopathy,other disorder>21 days;G2 uveitis/eye pain/blurred vision/decreased visual acuity;G4 other disorder.Decrease LVEF>10% G>=3 cardiac disorders.G3/4 hypertension.G3 fatigue>=7 days,hypersensitivity,infusion reaction,fever>=72 hours/hemodynamic compromise,endocrinopathy.G>=2 interstitial lung disease/pneumonitis;G3 bronchospasm.G3/4 rash,hand foot skin reaction,photosensitivity.G3 colitis;G3/4 diarrhea,nausea/vomiting.Neurologic G3.Other hematologic/nonhematolic G>=3 AE.

  2. Phase 2: Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v) 1.1 [From start of the treatment until disease progression, death or initiation of new anticancer therapy, whichever occurred first (Phase 2: maximum up to 26 months approximately)]

    ORR was defined as the percentage of participants who achieved a best overall response (BOR) of complete response (CR) or partial response (PR) as determined by investigator per RECIST v1.1. As per RECIST v1.1, CR was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes must have short axis measures less than (<)10 millimeter (mm). PR was defined as at least a 30% decrease in the sum of measures (longest diameter for tumor lesions and short axis measure for nodes) of target lesions, taking as reference baseline sum of diameters. Non-target lesions must be non-progressive disease (PD). PD was defined as at least a 20% increase in sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions was considered progression.

Secondary Outcome Measures

  1. Phase 1b: Objective Response Rate (ORR) Per RECIST v1.1 [From start of the treatment until disease progression, death or initiation of new anticancer therapy, whichever occurred first (Phase 1b: maximum up to 9 months approximately)]

    ORR was defined as the percentage of participants who achieved a BOR of CR or PR as determined by investigator per RECIST v1.1. As per RECIST v1.1, CR was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes must have short axis measures <10 mm. PR was defined as at least a 30% decrease in the sum of measures (longest diameter for tumor lesions and short axis measure for nodes) of target lesions, taking as reference the baseline sum of diameters. Non-target lesions must be non-PD. PD was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions was also considered progression.

  2. Duration of Response (DOR) as Per RECIST v1.1 [From date of first documented CR/PR to date of first documented PD, death or initiation of new anticancer therapy, whichever occurred first (Phase 1b: maximum up to 9 months approximately, Phase 2: maximum up to 26 months approximately)]

    DOR was defined as the time between the date of the first documented confirmed response (PR or CR) and the date of the first documented progression or death due to any cause. As per RECIST v1.1: CR was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes short axis measures <10 mm. PR was defined as at least 30% decrease in sum of measures (tumor lesions- longest diameter and nodes- short axis) of target lesions, taking as reference baseline sum of diameters. PD was defined as at least 20% increase in sum of diameters of measured lesions taking as references smallest sum of diameters recorded on study (including baseline) and an absolute increase of >=5 mm or appearance of at least 1 new lesion. Unequivocal progression of existing non-target lesions.

  3. Percentage of Participants With Complete Response as Per RECIST v1.1 [From start of the treatment until disease progression, death or initiation of new anticancer therapy, whichever occurred first (Phase 1b: maximum up to 9 months approximately and Phase 2: maximum up to 26 months approximately)]

    Complete response as per RECIST v1.1 was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes must have short axis measures <10 mm.

  4. Number of Participants With Treatment-Emergent Adverse Events (TEAE) Based on Common Terminology Criteria for Adverse Events (CTCAE) v4.03 [From start of the treatment up to 30 days after last dose or start of new anticancer therapy minus 1 day, whichever occurred first (Phase 1b: maximum up to 9 months approximately and Phase 2: maximum up to 26 months approximately)]

    AE: any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. SAE: AE resulting in any of following outcomes/deemed significant for any other reason: death; initial /prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. TEAEs: events between first dose of study drug and up to 30 days after last dose or before start of new anticancer therapy minus 1 day, whichever occurred first. TEAE graded by CTCAE grade 4.03: Grade 3: severe/medically significant but not immediately life-threatening, hospitalization or prolongation of existing hospitalization indicated, disabling, limiting self-care activities of daily living (ADL); Grade 4: life-threatening consequence, urgent intervention indicated. In this outcome measure, number of participants with 'all grades' and 'Grade 3/4' were reported.

  5. Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on Common Terminology Criteria for Adverse Events (CTCAE) v4.03: Hematology and Coagulation [Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)]

    Hematological parameters included: Hemoglobin graded high, Hemoglobin graded low, Platelets count graded low, White Blood Cell (WBC) graded high, WBC graded low, Neutrophils graded low, Lymphocytes graded high, Lymphocytes graded low. Coagulation parameters included: International Normalized Ratio (INR) graded high, Activated Partial Thromboplastin Time (aPTT) graded high. Test abnormalities were graded by CTCAE v4.03 as Grade 1=mild; Grade 2=moderate; Grade 3/Grade 4=severe/life-threatening. A grade 0 was assigned for all non-missing values not graded as 1 or higher. If value was graded >=1 but falls within the normal range, the grade was reset to 0. Categories with at least 1 non-zero data values are reported.

  6. Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on Normal Range: Hematology and Coagulation [Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)]

    Hematology parameters: Basophils, Eosinophils, Hematocrit, Monocytes, Red blood cells (RBC). Coagulation parameters: Prothrombin Time. Laboratory values were as per laboratory normal ranges. Values above range were reported as high and values below range as low. Laboratory parameters were graded based on laboratory normal ranges as low, normal, high and missing are reported in this outcome measure. Categories with at least 1 non-zero data values are reported.

  7. Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on CTCAE v4.03: Chemistry [Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)]

    Abnormalities: Albumin graded low, Alkaline phosphatase (ALP) graded high, Alanine aminotransferase (ALT) graded high, Aspartate aminotransferase (AST) graded high, Bilirubin graded high, Amylase graded high, Creatinine graded high, Corrected calcium graded high, Creatine Kinase (CK) graded high, Glucose graded high, Glucose graded low, Lipase graded high, Magnesium graded high, Magnesium graded low, Potassium graded high, Potassium graded low, Sodium graded high and Sodium graded low. Test abnormalities were graded by CTCAE v4.03 as Grade 1=mild; Grade 2=moderate; Grade 3/Grade 4=severe/life-threatening. A grade 0 was assigned for all non-missing values not graded as 1 or higher. If value was graded >=1 but falls within the normal range, the grade was reset to 0. Categories with at least 1 non-zero data values are reported.

  8. Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on Normal Range: Chemistry and Thyroid Function [Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)]

    Chemistry laboratory parameters: Blood urea nitrogen (BUN), Total protein, Chloride, Cancer antigen 19-9 (CA 19-9), Brain natriuretic peptide (BNP), Bicarbonate, Direct bilirubin, Carcinoembryonic antigen (CEA), Lactate dehydrogenase (LDH), Uric acid, Troponin I. Thyroid panel laboratory parameters: Thyroid-stimulating hormone (TSH), Free triiodothyronine (T3), Free thyroxine (T4). Laboratory values were as per laboratory normal ranges. Values above range were reported as high and values below range as low. Shift in chemistry and thyroid panel severity from baseline grade low, normal, high and missing to the post baseline grades as low, normal, high and missing are reported in this outcome measure. Categories with at least 1 non-zero data values are reported.

  9. Number of Participants With Abnormal Hepatic Laboratory Values [Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)]

    Criteria for abnormal hepatic laboratory parameters: Aspartate aminotransferase (AST) and Alanine aminotransferase (ALT): >3* upper limit of normal (ULN), >5*ULN, >8*ULN, >10*ULN, >20*ULN; Total bilirubin (TBL) >1.5*ULN, >2*ULN; Alkaline phosphatase (ALP) >2*ULN, >3*ULN. Categories with at least 1 non-zero data values are reported.

  10. Concentration Versus Time Summary of Plasma Concentration of Binimetinib [1.5 hours post dose of binimetinib on Day 1, 15 of Cycle 1; pre dose of binimetinib on Day 15 of Cycle 1, 2, 3, 4, 5]

Eligibility Criteria

Criteria

Ages Eligible for Study:
18 Years and Older
Sexes Eligible for Study:
All
Accepts Healthy Volunteers:
No

Key Inclusion Criteria

  • Measurable, histologically/cytologically confirmed metastatic colorectal cancer (mCRC).

  • Able to provide a sufficient amount of representative tumor specimen for central laboratory testing of RAS mutation status and microsatellite stable (MSS).

  • If a fresh tissue sample is provided, a blood sample is required.

  • Metastatic colorectal cancer (mCRC) categorized as microsatellite stable (MSS) by polymerase chain reaction (PCR) per local assay at any time prior to Screening or by the central laboratory.

  • RAS mutation per local assay at any time prior to Screening or by the central laboratory.

  • Have received at least 1 prior line of therapy and meets at least one of the following criteria:

  • were unable to tolerate the prior first-line regimen

  • experienced disease progression during or after prior first-line regimen for metastatic disease

  • progressed during or within 3 months of completing adjuvant chemotherapy. Note: Generally, treatments that are separated by an event of progression are considered different regimens.

  • Have received no more than 2 prior lines of therapy (maintenance therapy given in the metastatic setting will not be considered a separate regimen). Generally, treatments that are separated by an event of progression are considered different regimens.

  • Adequate bone marrow, cardiac, kidney and liver function

  • Able to take oral medications

  • Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 0 or 1.

  • Female patients are either postmenopausal for at least 1 year, are surgically sterile for at least 6 weeks, or must agree to take appropriate precautions to avoid pregnancy from screening through follow-up if of child-bearing potential

  • Non-sterile male patients who are sexually active with female partners of childbearing potential must agree to follow instructions for acceptable or highly effective method(s) of contraception for the duration of study treatment and for 7 months after the last dose of study treatment with nivolumab

Key Exclusion Criteria

  • Prior treatment with any MEK inhibitor, an anti-PD-1, anti-PD-L1, anti-PD-L2, anti-CD137, or anti-CTLA-4 antibody, or any other antibody or drug specifically targeting T-cell co-stimulation or checkpoint pathways.

  • Any untreated central nervous system (CNS) lesion.

  • Patients with an active, known or suspected autoimmune disease. Patients with type I diabetes mellitus, hypothyroidism only requiring hormone replacement, skin disorders (such as vitiligo, psoriasis, or alopecia) not requiring systemic treatment, or conditions not expected to recur in the absence of an external trigger are permitted to enroll.

  • Known history of retinal vein occlusion (RVO).

  • Known history of Gilbert's syndrome.

  • Pregnant or breastfeeding females.

  • Treatment with systemic immunosuppressive medications (including but not limited to prednisone, cyclophosphamide, azathioprine, methotrexate, thalidomide, and anti-tumor necrosis factor [anti-TNF] agents) within 2 weeks prior to first day of study treatment:

  • History of thromboembolic or cerebrovascular events ≤ 6 months prior to starting study treatment, including transient ischemic attacks, cerebrovascular accidents, deep vein thrombosis or pulmonary emboli.

  • Uncontrolled hypertension defined as persistent systolic blood pressure ≥ 150 mmHg or diastolic blood pressure ≥ 100 mmHg despite current therapy.

  • Concurrent neuromuscular disorder that is associated with the potential of elevated creatine kinase (CK) (e.g., inflammatory myopathies, muscular dystrophy, amyotrophic lateral sclerosis, spinal muscular atrophy).

  • 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).

  • Known history of positive test for human immunodeficiency virus (HIV) or known acquired immunodeficiency syndrome (AIDS). NOTE: Testing for HIV must be performed at sites where mandated locally.

  • Any positive test for hepatitis B virus or hepatitis C virus indicating acute or chronic infection, and/or detectable virus.

Contacts and Locations

Locations

Site City State Country Postal Code
1 UCLA Hematology/Oncology Los Angeles California United States 90095
2 UCLA Hematology/Oncology - Santa Monica Santa Monica California United States 90404
3 Christiana Care Health Services, Helen F. Graham Cancer Center Pharmacy, Suite 3200 Newark Delaware United States 19713
4 Christiana Care Health Services, Helen F. Graham Cancer Center Newark Delaware United States 19713
5 Christiana Care Oncology Hematology, Helen F Graham Cancer Center, Suite 2400 Newark Delaware United States 19713
6 Medical Oncology Hematology Consultants, PA, Helen F. Graham Cancer Center Newark Delaware United States 19713
7 Christiana Care Health Services, Christiana Hospital Newark Delaware United States 19718
8 Georgetown University Medical Center Department of Pharmacy, Research Washington District of Columbia United States 20007
9 Georgetown University Medical Center Washington District of Columbia United States 20007
10 Hematology Oncology Associates of the Treasure Coast Port Saint Lucie Florida United States 34952
11 Indiana CTSI Clinical Research Center (ICRC) Indianapolis Indiana United States 46202
12 Indiana University Health Hospital Indianapolis Indiana United States 46202
13 Indiana University Health Melvin and Bren Simon Cancer Center Indianapolis Indiana United States 46202
14 Investigational Drug Services IUHSCC Indianapolis Indiana United States 46202
15 Sidney &Lois Eskenazi Hospital Indianapolis Indiana United States 46202
16 Spring Mill Medical Center Indianapolis Indiana United States 46290
17 Siteman Cancer Center - West County Creve Coeur Missouri United States 63141
18 Siteman Cancer Center - North County Florissant Missouri United States 63031
19 Center for Outpatient Health (Dermatology Clinic) Saint Louis Missouri United States 63108
20 Center for Outpatient Health (Ophthalmology Clinic) Saint Louis Missouri United States 63108
21 Barnes-Jewish Hospital Saint Louis Missouri United States 63110
22 Washington University School of Medicine Saint Louis Missouri United States 63110
23 Siteman Cancer Center - South County Saint Louis Missouri United States 63129
24 Siteman Cancer Center - Barnes St. Peters Saint Peters Missouri United States 63376
25 Hospital of the University of Pennsylvania Philadelphia Pennsylvania United States 19104
26 Investigational Drug Service of the University of Pennsylvania Philadelphia Pennsylvania United States 19104
27 Fox Chase Cancer Center Philadelphia Pennsylvania United States 19111
28 Sarah Cannon Research Institute Chattanooga Tennessee United States 37404
29 SCRI Tennessee Oncology Chattanooga Chattanooga Tennessee United States 37404
30 Tennessee Oncology, PLLC Cleveland Tennessee United States 37311
31 Tennessee Oncology NASH - SCRI - PPDS Nashville Tennessee United States 37203
32 The Sarah Cannon Research Institute. Nashville Tennessee United States 37203
33 The University of Texas MD Anderson Cancer Center Houston Texas United States 77030
34 UZ Leuven - Dermatology Leuven Belgium 3000
35 UZ Leuven - Ophthalmology Leuven Belgium 3000
36 UZ Leuven Leuven Belgium 3000
37 The Netherlands Cancer Institute Antoni Van Leeuwenhoek Amsterdam Noord-holland Netherlands 1066 CX
38 OLVG locatie Oost Amsterdam Noord-holland Netherlands 1091 AC
39 Amsterdam Medical Center (AMC) Amsterdam Noord-holland Netherlands 1105 AZ
40 Hospital Universitario Vall d'Hebron Barcelona Spain 08035
41 Hospital Universitario Vall d'Hebrón - PPDS Barcelona Spain 08035
42 Hospital General Universitario Gregorio Marañon Madrid Spain 28007
43 Clinica Rementeria MAdrid Spain 28010
44 Hospital Universitario 12 Octubre Madrid Spain 28041
45 Hospital HM Universitario Sanchinarro, CIOCC Madrid Spain 28050
46 Royal Marsden Hospital - London London London, CITY OF United Kingdom SW3 6JJ
47 Churchill Hospital Oxford Oxfordshire United Kingdom OX3 7LE
48 Royal Marsden Hospital NHS Foundation Trust Surrey Sutton United Kingdom SM2 5PT
49 Royal Marsden Hospital NHS Foundation Trust London United Kingdom SW3 6JJ
50 Royal Marsden Hospital -Fulham Road London United Kingdom SW36JJ

Sponsors and Collaborators

  • Pfizer
  • Bristol-Myers Squibb

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:
NCT03271047
Other Study ID Numbers:
  • ARRAY-162-202
  • C4211004
  • 2017-003464-12
First Posted:
Sep 1, 2017
Last Update Posted:
Jan 4, 2022
Last Verified:
Dec 1, 2021
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
Pre-assignment Detail This study included 2 phases: Phase 1b and Phase 2. The recommended dose for Phase 2 (RP2D) of binimetinib was determined in Phase 1b (dose finding phase).
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated microsatellite-stable (MSS) metastatic colorectal cancer with rat sarcoma virus (RAS) mutation received binimetinib at a starting dose of 45 milligrams (mg) tablet orally twice daily (BID) along with 480 mg intravenously (IV) dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 milligram per kilogram (mg/kg) IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Period Title: Phase 1b: Dose Finding Period
STARTED 10 11 0 0
COMPLETED 0 0 0 0
NOT COMPLETED 10 11 0 0
Period Title: Phase 1b: Dose Finding Period
STARTED 0 0 27 27
COMPLETED 0 0 0 0
NOT COMPLETED 0 0 27 27

Baseline Characteristics

Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib Total
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Total of all reporting groups
Overall Participants 10 11 27 27 75
Age (Count of Participants)
<=18 years
0
0%
0
0%
0
0%
0
0%
0
0%
Between 18 and 65 years
5
50%
9
81.8%
19
70.4%
20
74.1%
53
70.7%
>=65 years
5
50%
2
18.2%
8
29.6%
7
25.9%
22
29.3%
Sex: Female, Male (Count of Participants)
Female
2
20%
4
36.4%
11
40.7%
10
37%
27
36%
Male
8
80%
7
63.6%
16
59.3%
17
63%
48
64%
Ethnicity (NIH/OMB) (Count of Participants)
Hispanic or Latino
0
0%
0
0%
1
3.7%
0
0%
1
1.3%
Not Hispanic or Latino
10
100%
11
100%
21
77.8%
23
85.2%
65
86.7%
Unknown or Not Reported
0
0%
0
0%
5
18.5%
4
14.8%
9
12%
Race (NIH/OMB) (Count of Participants)
American Indian or Alaska Native
0
0%
0
0%
0
0%
0
0%
0
0%
Asian
0
0%
0
0%
1
3.7%
0
0%
1
1.3%
Native Hawaiian or Other Pacific Islander
0
0%
0
0%
0
0%
0
0%
0
0%
Black or African American
1
10%
0
0%
1
3.7%
1
3.7%
3
4%
White
9
90%
11
100%
21
77.8%
23
85.2%
64
85.3%
More than one race
0
0%
0
0%
0
0%
0
0%
0
0%
Unknown or Not Reported
0
0%
0
0%
4
14.8%
3
11.1%
7
9.3%

Outcome Measures

1. Primary Outcome
Title Phase 1b: Number of Participants With Dose-Limiting Toxicities (DLT)
Description DLT:Adverse event(AE)/abnormal laboratory assessed unrelated-disease,disease progression,intercurrent illness/concomitant medication/therapies resulting inability tolerate 75% dose intensity in Cycle 1.Total bilirubin(TBL)grade(G)>=3 (>3.0*upper limit of normal[ULN)]);AST/ALT>5-8*ULN>5 days,>8*ULN,>3*ULN concurrent TBL>2*ULN;G>=3 serum creatinine,CK elevation,ECG QTcF prolonged,G3 troponin,electrolyte>72 hours,G3/4 amylase/lipase.G4 ANC,platelet count>7 days;G3/4 platelet count,other AE except lymphopenia.G>=3 retinopathy,other disorder>21 days;G2 uveitis/eye pain/blurred vision/decreased visual acuity;G4 other disorder.Decrease LVEF>10% G>=3 cardiac disorders.G3/4 hypertension.G3 fatigue>=7 days,hypersensitivity,infusion reaction,fever>=72 hours/hemodynamic compromise,endocrinopathy.G>=2 interstitial lung disease/pneumonitis;G3 bronchospasm.G3/4 rash,hand foot skin reaction,photosensitivity.G3 colitis;G3/4 diarrhea,nausea/vomiting.Neurologic G3.Other hematologic/nonhematolic G>=3 AE.
Time Frame Cycle 1: Day 1 up to Day 28

Outcome Measure Data

Analysis Population Description
Dose-Determining Set (DDS) included all participants in Phase 1b who experienced a DLT or receive at least 75% of the planned binimetinib dose intensity during Cycle 1.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days.
Measure Participants 9 11
Count of Participants [Participants]
1
10%
2
18.2%
2. Primary Outcome
Title Phase 2: Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v) 1.1
Description ORR was defined as the percentage of participants who achieved a best overall response (BOR) of complete response (CR) or partial response (PR) as determined by investigator per RECIST v1.1. As per RECIST v1.1, CR was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes must have short axis measures less than (<)10 millimeter (mm). PR was defined as at least a 30% decrease in the sum of measures (longest diameter for tumor lesions and short axis measure for nodes) of target lesions, taking as reference baseline sum of diameters. Non-target lesions must be non-progressive disease (PD). PD was defined as at least a 20% increase in sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions was considered progression.
Time Frame From start of the treatment until disease progression, death or initiation of new anticancer therapy, whichever occurred first (Phase 2: maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
All participants randomized to study treatment in Phase 2.
Arm/Group Title Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 27 27
Number (95% Confidence Interval) [Percentage of participants]
0.0
0%
7.4
67.3%
3. Secondary Outcome
Title Phase 1b: Objective Response Rate (ORR) Per RECIST v1.1
Description ORR was defined as the percentage of participants who achieved a BOR of CR or PR as determined by investigator per RECIST v1.1. As per RECIST v1.1, CR was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes must have short axis measures <10 mm. PR was defined as at least a 30% decrease in the sum of measures (longest diameter for tumor lesions and short axis measure for nodes) of target lesions, taking as reference the baseline sum of diameters. Non-target lesions must be non-PD. PD was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions was also considered progression.
Time Frame From start of the treatment until disease progression, death or initiation of new anticancer therapy, whichever occurred first (Phase 1b: maximum up to 9 months approximately)

Outcome Measure Data

Analysis Population Description
All participants who received at least 1 dose of any study treatment in Phase 1b.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days.
Measure Participants 10 11
Number (95% Confidence Interval) [Percentage of participants]
0.0
0%
0.0
0%
4. Secondary Outcome
Title Duration of Response (DOR) as Per RECIST v1.1
Description DOR was defined as the time between the date of the first documented confirmed response (PR or CR) and the date of the first documented progression or death due to any cause. As per RECIST v1.1: CR was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes short axis measures <10 mm. PR was defined as at least 30% decrease in sum of measures (tumor lesions- longest diameter and nodes- short axis) of target lesions, taking as reference baseline sum of diameters. PD was defined as at least 20% increase in sum of diameters of measured lesions taking as references smallest sum of diameters recorded on study (including baseline) and an absolute increase of >=5 mm or appearance of at least 1 new lesion. Unequivocal progression of existing non-target lesions.
Time Frame From date of first documented CR/PR to date of first documented PD, death or initiation of new anticancer therapy, whichever occurred first (Phase 1b: maximum up to 9 months approximately, Phase 2: maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
FAS included all participants who received at least 1 dose of any study drug in Phase 1b and all participants randomized to study treatment in Phase 2. Here, "Overall Number of Participants Analyzed" signifies number of participants who achieved an objective response. There were no participants who had event in Phase 1b and Nivolumab and Binimetinib group of Phase 2.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 0 0 0 2
Median (95% Confidence Interval) [Months]
11.4
5. Secondary Outcome
Title Percentage of Participants With Complete Response as Per RECIST v1.1
Description Complete response as per RECIST v1.1 was defined as disappearance of target and non-target lesions and normalization of tumor markers. Pathological lymph nodes must have short axis measures <10 mm.
Time Frame From start of the treatment until disease progression, death or initiation of new anticancer therapy, whichever occurred first (Phase 1b: maximum up to 9 months approximately and Phase 2: maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
FAS included all participants who received at least 1 dose of any study drug in Phase 1b and all participants randomized to study treatment in Phase 2.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 27 27
Number [Percentage of participants]
0
0%
0
0%
0
0%
0
0%
6. Secondary Outcome
Title Number of Participants With Treatment-Emergent Adverse Events (TEAE) Based on Common Terminology Criteria for Adverse Events (CTCAE) v4.03
Description AE: any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. SAE: AE resulting in any of following outcomes/deemed significant for any other reason: death; initial /prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. TEAEs: events between first dose of study drug and up to 30 days after last dose or before start of new anticancer therapy minus 1 day, whichever occurred first. TEAE graded by CTCAE grade 4.03: Grade 3: severe/medically significant but not immediately life-threatening, hospitalization or prolongation of existing hospitalization indicated, disabling, limiting self-care activities of daily living (ADL); Grade 4: life-threatening consequence, urgent intervention indicated. In this outcome measure, number of participants with 'all grades' and 'Grade 3/4' were reported.
Time Frame From start of the treatment up to 30 days after last dose or start of new anticancer therapy minus 1 day, whichever occurred first (Phase 1b: maximum up to 9 months approximately and Phase 2: maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
The safety set included of all participants who received at least 1 dose of study drug.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 27 27
Treatment emergent AEs- All grades
10
100%
11
100%
27
100%
27
100%
Treatment emergent SAEs- All grades
5
50%
6
54.5%
12
44.4%
11
40.7%
Treatment emergent AEs- Grade 3/4
6
60%
8
72.7%
19
70.4%
21
77.8%
Treatment emergent SAEs- Grade 3/4
5
50%
6
54.5%
11
40.7%
10
37%
7. Secondary Outcome
Title Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on Common Terminology Criteria for Adverse Events (CTCAE) v4.03: Hematology and Coagulation
Description Hematological parameters included: Hemoglobin graded high, Hemoglobin graded low, Platelets count graded low, White Blood Cell (WBC) graded high, WBC graded low, Neutrophils graded low, Lymphocytes graded high, Lymphocytes graded low. Coagulation parameters included: International Normalized Ratio (INR) graded high, Activated Partial Thromboplastin Time (aPTT) graded high. Test abnormalities were graded by CTCAE v4.03 as Grade 1=mild; Grade 2=moderate; Grade 3/Grade 4=severe/life-threatening. A grade 0 was assigned for all non-missing values not graded as 1 or higher. If value was graded >=1 but falls within the normal range, the grade was reset to 0. Categories with at least 1 non-zero data values are reported.
Time Frame Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
Safety set included of all participants who received at least 1 dose of study drug. Here "Number Analyzed" signifies number of participants evaluable for specified rows.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 27 27
Hemoglobin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
3
30%
3
27.3%
4
14.8%
5
18.5%
Hemoglobin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
2
20%
2
18.2%
6
22.2%
10
37%
Hemoglobin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 2 (at post baseline)
2
20%
1
9.1%
2
7.4%
1
3.7%
Hemoglobin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Hemoglobin- CTCAE Graded Low: Grade 1 (at baseline) to Grade 0 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Hemoglobin- CTCAE Graded Low: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
4
36.4%
4
14.8%
4
14.8%
Hemoglobin- CTCAE Graded Low: Grade 1 (at baseline) to Grade 2 (at post baseline)
2
20%
0
0%
3
11.1%
1
3.7%
Hemoglobin- CTCAE Graded Low: Grade 1 (at baseline) to Grade 3 (at post baseline)
0
0%
1
9.1%
1
3.7%
1
3.7%
Hemoglobin- CTCAE Graded Low: Grade 1 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Hemoglobin- CTCAE Graded Low: Grade 2 (at baseline) to Grade 2 (at post baseline)
3
30%
5
45.5%
Hemoglobin- CTCAE Graded Low: Grade 2 (at baseline) to Grade 3 (at post baseline)
1
10%
0
0%
Hemoglobin- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
10
100%
11
100%
26
96.3%
26
96.3%
Hemoglobin- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Hemoglobin- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Lymphocytes - CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
6
60%
7
63.6%
11
40.7%
11
40.7%
Lymphocytes- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
2
7.4%
4
14.8%
Lymphocytes- CTCAE Graded Low: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
2
18.2%
4
14.8%
6
22.2%
Lymphocytes- CTCAE Graded Low: Grade 0 (at baseline) to Grade 3 (at post baseline)
1
10%
0
0%
3
11.1%
1
3.7%
Lymphocytes- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Lymphocytes- CTCAE Graded Low: Grade 1 (at baseline) to Grade 0 (at post baseline)
0
0%
0
0%
1
3.7%
Lymphocytes- CTCAE Graded Low: Grade 1 (at baseline) to Grade 2 (at post baseline)
1
10%
1
9.1%
0
0%
Lymphocytes- CTCAE Graded Low: Grade 1 (at baseline) to Grade 3 (at post baseline)
0
0%
1
9.1%
0
0%
Lymphocytes- CTCAE Graded Low: Grade 2 (at baseline) to Grade 0 (at post baseline)
0
0%
1
9.1%
0
0%
Lymphocytes- CTCAE Graded Low: Grade 2 (at baseline) to Grade 2 (at post baseline)
1
10%
1
9.1%
2
7.4%
Lymphocytes- CTCAE Graded Low: Grade 2 (at baseline) to Grade 3 (at post baseline)
1
10%
1
9.1%
2
7.4%
Lymphocytes- CTCAE Graded Low: Grade 3 (at baseline) to Grade 0 (at post baseline)
0
0%
0
0%
1
3.7%
Lymphocytes- CTCAE Graded Low: Grade 3 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
0
0%
Lymphocytes- CTCAE Graded Low: Grade 3 (at baseline) to Grade 3 (at post baseline)
1
10%
0
0%
0
0%
Lymphocytes- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
9
90%
11
100%
25
92.6%
26
96.3%
Lymphocytes- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
1
10%
0
0%
1
3.7%
1
3.7%
Lymphocytes- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Neutrophils- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
9
90%
11
100%
23
85.2%
26
96.3%
Neutrophils- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
1
3.7%
1
3.7%
Neutrophils- CTCAE Graded Low: Grade 0 (at baseline) to Grade 2 (at post baseline)
1
10%
0
0%
0
0%
0
0%
Neutrophils- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Neutrophils- CTCAE Graded Low: Grade 1 (at baseline) to Grade 0 (at post baseline)
1
10%
Neutrophils- CTCAE Graded Low: Grade 1 (at baseline) to Grade 3 (at post baseline)
1
10%
Platelets- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
8
80%
9
81.8%
21
77.8%
22
81.5%
Platelets- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
2
20%
1
9.1%
3
11.1%
4
14.8%
Platelets- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
2
7.4%
0
0%
Platelets- CTCAE Graded Low: Grade 1 (at baseline) to Grade 0 (at post baseline)
1
10%
0
0%
0
0%
Platelets- CTCAE Graded Low: Grade 1 (at baseline) to Grade 1 (at post baseline)
0
0%
1
9.1%
1
3.7%
WBC- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
9
90%
11
100%
21
77.8%
24
88.9%
WBC- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
2
7.4%
0
0%
WBC- CTCAE Graded Low: Grade 0 (at baseline) to Grade 2 (at post baseline)
1
10%
0
0%
1
3.7%
2
7.4%
WBC- CTCAE Graded Low: Grade 0 (at baseline) to Grade 4 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
WBC- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
WBC- CTCAE Graded Low: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
0
0%
WBC- CTCAE Graded Low: Grade 1 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
WBC- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
10
100%
11
100%
26
96.3%
27
100%
WBC- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
aPTT- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
6
60%
8
72.7%
16
59.3%
17
63%
aPTT- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
3
30%
2
18.2%
6
22.2%
6
22.2%
aPTT- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
1
10%
0
0%
2
7.4%
1
3.7%
aPTT- CTCAE Graded High: Grade 1 (at baseline) to Grade 0 (at post baseline)
0
0%
0
0%
1
3.7%
aPTT- CTCAE Graded High: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
1
9.1%
1
3.7%
aPTT- CTCAE Graded High: Grade 1 (at baseline) to Grade 3 (at post baseline)
0
0%
1
9.1%
0
0%
aPTT- CTCAE Graded High: Grade 1 (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
0
0%
aPTT- CTCAE Graded High: Missing (at baseline) to Grade 0 (at post baseline)
1
10%
INR- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
5
50%
6
54.5%
16
59.3%
13
48.1%
INR- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
4
40%
2
18.2%
4
14.8%
4
14.8%
INR- CTCAE Graded High: Grade 1 (at baseline) to Grade 0 (at post baseline)
0
0%
1
9.1%
1
3.7%
1
3.7%
INR- CTCAE Graded High: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
1
9.1%
5
18.5%
8
29.6%
INR- CTCAE Graded High: Grade 1 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
INR- CTCAE Graded High: Grade 2 (at baseline) to Grade 2 (at post baseline)
1
10%
INR- CTCAE Graded High: Missing (at baseline) to Grade 0 (at post baseline)
1
10%
8. Secondary Outcome
Title Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on Normal Range: Hematology and Coagulation
Description Hematology parameters: Basophils, Eosinophils, Hematocrit, Monocytes, Red blood cells (RBC). Coagulation parameters: Prothrombin Time. Laboratory values were as per laboratory normal ranges. Values above range were reported as high and values below range as low. Laboratory parameters were graded based on laboratory normal ranges as low, normal, high and missing are reported in this outcome measure. Categories with at least 1 non-zero data values are reported.
Time Frame Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
Safety set included of all participants who received at least 1 dose of study drug. Here "Number Analyzed" signifies number of participants evaluable for specified rows.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 27 27
Basophils: Normal (at baseline) to Low (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Basophils: Normal (at baseline) to Normal (at post baseline)
10
100%
11
100%
25
92.6%
27
100%
Basophils: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Eosinophils: Normal (at baseline) to Low (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Eosinophils: Normal (at baseline) to Normal (at post baseline)
7
70%
11
100%
25
92.6%
20
74.1%
Eosinophils: Normal (at baseline) to High (at post baseline)
2
20%
0
0%
1
3.7%
6
22.2%
Eosinophils: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Eosinophils: High (at baseline) to Normal (at post baseline)
1
10%
Hematocrit: Low (at baseline) to Low (at post baseline)
2
20%
1
9.1%
8
29.6%
9
33.3%
Hematocrit: Low (at baseline) to Normal (at post baseline)
0
0%
1
9.1%
1
3.7%
0
0%
Hematocrit: Low (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Hematocrit: Normal (at baseline) to Low (at post baseline)
5
50%
4
36.4%
13
48.1%
12
44.4%
Hematocrit: Normal (at baseline) to Normal (at post baseline)
3
30%
4
36.4%
4
14.8%
6
22.2%
Hematocrit: Normal (at baseline) to High & Low (at post baseline)
0
0%
1
9.1%
0
0%
0
0%
Monocytes: Normal (at baseline) to Low (at post baseline)
1
10%
0
0%
1
3.7%
0
0%
Monocytes: Normal (at baseline) to Normal (at post baseline)
9
90%
7
63.6%
19
70.4%
20
74.1%
Monocytes: Normal (at baseline) to High (at post baseline)
0
0%
4
36.4%
6
22.2%
6
22.2%
Monocytes: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Monocytes: High (at baseline) to Normal (at post baseline)
1
10%
RBC: Low (at baseline) to Low (at post baseline)
1
10%
1
9.1%
13
48.1%
6
22.2%
RBC: Low (at baseline) to Normal (at post baseline)
0
0%
1
9.1%
1
3.7%
1
3.7%
RBC: Normal (at baseline) to Low (at post baseline)
6
60%
5
45.5%
8
29.6%
10
37%
RBC: Normal (at baseline) to Normal (at post baseline)
2
20%
4
36.4%
3
11.1%
9
33.3%
RBC: Normal (at baseline) to High (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
RBC: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
RBC: High (at baseline) to Low (at post baseline)
0
0%
1
9.1%
RBC: High (at baseline) to Normal (at post baseline)
1
10%
0
0%
Prothrombin Time: Normal (at baseline) to Normal (at post baseline)
3
30%
3
27.3%
8
29.6%
8
29.6%
Prothrombin Time: Normal (at baseline) to High (at post baseline)
4
40%
4
36.4%
11
40.7%
9
33.3%
Prothrombin Time: High (at baseline) to Normal (at post baseline)
0
0%
1
9.1%
0
0%
0
0%
Prothrombin Time: High (at baseline) to High (at post baseline)
3
30%
3
27.3%
7
25.9%
9
33.3%
Prothrombin Time: High (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Prothrombin Time: Missing (at baseline) to High (at post baseline)
1
10%
9. Secondary Outcome
Title Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on CTCAE v4.03: Chemistry
Description Abnormalities: Albumin graded low, Alkaline phosphatase (ALP) graded high, Alanine aminotransferase (ALT) graded high, Aspartate aminotransferase (AST) graded high, Bilirubin graded high, Amylase graded high, Creatinine graded high, Corrected calcium graded high, Creatine Kinase (CK) graded high, Glucose graded high, Glucose graded low, Lipase graded high, Magnesium graded high, Magnesium graded low, Potassium graded high, Potassium graded low, Sodium graded high and Sodium graded low. Test abnormalities were graded by CTCAE v4.03 as Grade 1=mild; Grade 2=moderate; Grade 3/Grade 4=severe/life-threatening. A grade 0 was assigned for all non-missing values not graded as 1 or higher. If value was graded >=1 but falls within the normal range, the grade was reset to 0. Categories with at least 1 non-zero data values are reported.
Time Frame Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
Safety set included of all participants who received at least 1 dose of study drug. Here "Number Analyzed" signifies number of participants evaluable for specified rows.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 27 27
ALT- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
5
50%
5
45.5%
12
44.4%
12
44.4%
ALT- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
3
30%
5
45.5%
7
25.9%
5
18.5%
ALT- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
0
0%
1
3.7%
1
3.7%
ALT- CTCAE Graded High: Grade 0 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
0
0%
3
11.1%
ALT- CTCAE Graded High: Grade 1 (at baseline) to Grade 0 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
ALT- CTCAE Graded High: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
1
9.1%
3
11.1%
2
7.4%
ALT- CTCAE Graded High: Grade 1 (at baseline) to Grade 2 (at post baseline)
0
0%
0
0%
1
3.7%
2
7.4%
ALT- CTCAE Graded High: Grade 1 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
ALT- CTCAE Graded High: Grade 2 (at baseline) to Grade 2 (at post baseline)
1
10%
1
9.1%
0
0%
ALT- CTCAE Graded High: Grade 2 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
1
3.7%
ALT- CTCAE Graded High: Grade 3 (at baseline) to Missing (at post baseline)
1
10%
AST- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
3
30%
2
18.2%
8
29.6%
5
18.5%
AST- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
5
50%
4
36.4%
11
40.7%
13
48.1%
AST- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
2
7.4%
2
7.4%
AST- CTCAE Graded High: Grade 0 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
0
0%
2
7.4%
AST- CTCAE Graded High: Grade 1 (at baseline) to Grade 1 (at post baseline)
2
20%
4
36.4%
3
11.1%
1
3.7%
AST- CTCAE Graded High: Grade 1 (at baseline) to Grade 2 (at post baseline)
0
0%
0
0%
1
3.7%
3
11.1%
AST- CTCAE Graded High: Grade 1 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
1
3.7%
1
3.7%
AST- CTCAE Graded High: Grade 2 (at baseline) to Missing (at post baseline)
1
10%
Albumin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
4
40%
2
18.2%
8
29.6%
8
29.6%
Albumin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
3
30%
2
18.2%
9
33.3%
11
40.7%
Albumin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 2 (at post baseline)
3
30%
4
36.4%
7
25.9%
6
22.2%
Albumin- CTCAE Graded Low: Grade 0 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Albumin- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Albumin- CTCAE Graded Low: Grade 1 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
1
3.7%
Albumin- CTCAE Graded Low: Grade 1 (at baseline) to Grade 2 (at post baseline)
2
20%
2
18.2%
0
0%
Albumin- CTCAE Graded Low: Grade 2 (at baseline) to Grade 2 (at post baseline)
1
10%
Alkaline phosphatase- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
2
20%
2
18.2%
7
25.9%
6
22.2%
Alkaline phosphatase- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
3
30%
1
9.1%
8
29.6%
5
18.5%
Alkaline phosphatase- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
0
0%
1
3.7%
Alkaline phosphatase- CTCAE Graded High: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
0
0%
5
18.5%
7
25.9%
Alkaline phosphatase- CTCAE Graded High: Grade 1 (at baseline) to Grade 2 (at post baseline)
2
20%
4
36.4%
3
11.1%
3
11.1%
Alkaline phosphatase- CTCAE Graded High: Grade 1 (at baseline) to Grade 3 (at post baseline)
1
10%
0
0%
1
3.7%
1
3.7%
Alkaline phosphatase- CTCAE Graded High: Grade 2 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
0
0%
0
0%
Alkaline phosphatase- CTCAE Graded High: Grade 2 (at baseline) to Grade 3 (at post baseline)
1
10%
0
0%
2
7.4%
2
7.4%
Alkaline phosphatase- CTCAE Graded High: Grade 3 (at baseline) to Grade 3 (at post baseline)
2
20%
0
0%
2
7.4%
Alkaline phosphatase- CTCAE Graded High: Grade 3 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Amylase- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
8
80%
7
63.6%
18
66.7%
20
74.1%
Amylase- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
1
10%
2
18.2%
5
18.5%
2
7.4%
Amylase- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
0
0%
1
3.7%
2
7.4%
Amylase- CTCAE Graded High: Grade 0 (at baseline) to Grade 3 (at post baseline)
0
0%
1
9.1%
2
7.4%
2
7.4%
Amylase- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
1
3.7%
0
0%
Amylase- CTCAE Graded High: Grade 1 (at baseline) to Grade 2 (at post baseline)
1
10%
Amylase- CTCAE Graded High: Grade 2 (at baseline) to Grade 4 (at post baseline)
1
10%
Bilirubin- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
10
100%
9
81.8%
25
92.6%
24
88.9%
Bilirubin- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
1
3.7%
2
7.4%
Bilirubin- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
0
0%
1
3.7%
Bilirubin- CTCAE Graded High: Grade 1 (at baseline) to Grade 2 (at post baseline)
1
10%
Bilirubin- CTCAE Graded High: Grade 4 (at baseline) to Missing (at post baseline)
1
10%
Corrected Calcium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
7
70%
11
100%
24
88.9%
25
92.6%
Corrected Calcium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
3
30%
0
0%
2
7.4%
1
3.7%
Corrected Calcium- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Corrected Calcium- CTCAE Graded Low: Grade 1 (at baseline) to Grade 0 (at post baseline)
1
10%
Corrected Calcium- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
10
100%
11
100%
25
92.6%
25
92.6%
Corrected Calcium- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
1
3.7%
1
3.7%
Corrected Calcium- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Corrected Calcium- CTCAE Graded High: Grade 1 (at baseline) to Grade 0 (at post baseline)
1
10%
Creatine kinase- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
1
10%
3
27.3%
3
11.1%
7
25.9%
Creatine kinase- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
3
30%
4
36.4%
11
40.7%
7
25.9%
Creatine kinase- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
4
40%
2
18.2%
6
22.2%
8
29.6%
Creatine kinase- CTCAE Graded High: Grade 0 (at baseline) to Grade 3 (at post baseline)
1
10%
1
9.1%
5
18.5%
1
3.7%
Creatine kinase- CTCAE Graded High: Grade 0 (at baseline) to Grade 4 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Creatine kinase- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Creatine kinase- CTCAE Graded High: Grade 1 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
1
3.7%
Creatine kinase- CTCAE Graded High: Grade 1 (at baseline) to Grade 3 (at post baseline)
1
10%
0
0%
1
3.7%
Creatine kinase- CTCAE Graded High: Grade 1 (at baseline) to Grade 4 (at post baseline)
0
0%
0
0%
1
3.7%
Creatine kinase - CTCAE Graded High: Missing (at baseline) to Grade 1 (at post baseline)
1
10%
Creatinine- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
1
10%
2
18.2%
6
22.2%
7
25.9%
Creatinine- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
7
70%
7
63.6%
14
51.9%
18
66.7%
Creatinine- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
2
20%
2
18.2%
5
18.5%
2
7.4%
Creatinine- CTCAE Graded High: Grade 0 (at baseline) to Grade 4 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Creatinine- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Glucose- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
10
100%
10
90.9%
23
85.2%
26
96.3%
Glucose- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
1
9.1%
1
3.7%
0
0%
Glucose- CTCAE Graded Low: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Glucose- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Glucose- CTCAE Graded Low: Grade 1 (at baseline) to Grade 0 (at post baseline)
0
0%
1
9.1%
Glucose- CTCAE Graded Low: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
0
0%
Glucose- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
7
70%
6
54.5%
18
66.7%
14
51.9%
Glucose- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
1
9.1%
1
3.7%
1
3.7%
Glucose - CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
1
10%
1
9.1%
0
0%
1
3.7%
Glucose- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
1
10%
2
18.2%
5
18.5%
5
18.5%
Glucose- CTCAE Graded High: Grade 1 (at baseline) to Grade 0 (at post baseline)
1
10%
0
0%
Glucose - CTCAE Graded High: Grade 1 (at baseline) to Grade 2 (at post baseline)
0
0%
1
9.1%
Glucose- CTCAE Graded High: Grade 3 (at baseline) to Grade 2 (at post baseline)
1
10%
0
0%
Glucose- CTCAE Graded High: Grade 3 (at baseline) to Grade 3 (at post baseline)
0
0%
1
9.1%
Glucose- CTCAE Graded High: Grade 3 (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
Glucose- CTCAE Graded High: Missing (at baseline) to Grade 0 (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Glucose- CTCAE Graded High: Missing (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Glucose- CTCAE Graded High: Missing (at baseline) to Missing (at post baseline)
1
10%
1
9.1%
0
0%
2
7.4%
Lipase- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
5
50%
6
54.5%
17
63%
15
55.6%
Lipase- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
2
20%
2
18.2%
2
7.4%
4
14.8%
Lipase- CTCAE Graded High: Grade 0 (at baseline) to Grade 2 (at post baseline)
1
10%
1
9.1%
1
3.7%
0
0%
Lipase- CTCAE Graded High: Grade 0 (at baseline) to Grade 3 (at post baseline)
1
10%
0
0%
4
14.8%
3
11.1%
Lipase- CTCAE Graded High: Grade 0 (at baseline) to Grade 4 (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Lipase- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
1
3.7%
0
0%
Lipase- CTCAE Graded High: Grade 1 (at baseline) to Grade 0 (at post baseline)
1
10%
0
0%
1
3.7%
0
0%
Lipase- CTCAE Graded High: Grade 1 (at baseline) to Grade 1 (at post baseline)
0
0%
1
9.1%
1
3.7%
2
7.4%
Lipase- CTCAE Graded High: Grade 1 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Lipase- CTCAE Graded High: Grade 4 (at baseline) to Grade 4 (at post baseline)
1
10%
Magnesium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
8
80%
10
90.9%
22
81.5%
24
88.9%
Magnesium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
1
10%
1
9.1%
3
11.1%
2
7.4%
Magnesium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 2 (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Magnesium- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Magnesium- CTCAE Graded Low: Grade 1 (at baseline) to Grade 2 (at post baseline)
1
10%
Magnesium- CTCAE Graded Low: Grade 2 (at baseline) to Grade 3 (at post baseline)
1
10%
Magnesium- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
10
100%
11
100%
26
96.3%
27
100%
Magnesium- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Potassium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
7
70%
9
81.8%
20
74.1%
23
85.2%
Potassium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
3
30%
2
18.2%
3
11.1%
4
14.8%
Potassium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Potassium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 4 (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Potassium- CTCAE Graded Low: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Potassium- CTCAE Graded Low: Grade 3 (at baseline) to Grade 3 (at post baseline)
1
10%
Potassium- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
9
90%
10
90.9%
23
85.2%
23
85.2%
Potassium- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
1
10%
1
9.1%
2
7.4%
4
14.8%
Potassium- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Potassium- CTCAE Graded High: Grade 1 (at baseline) to Grade 0 (at post baseline)
1
10%
Sodium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 0 (at post baseline)
9
90%
7
63.6%
19
70.4%
23
85.2%
Sodium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 1 (at post baseline)
1
10%
3
27.3%
3
11.1%
1
3.7%
Sodium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 3 (at post baseline)
0
0%
0
0%
1
3.7%
2
7.4%
Sodium- CTCAE Graded Low: Grade 0 (at baseline) to Grade 4 (at post baseline)
0
0%
0
0%
2
7.4%
1
3.7%
Sodium- CTCAE Graded Low: Grade 1 (at baseline) to Grade 1 (at post baseline)
1
10%
Sodium- CTCAE Graded Low: Grade 1 (at baseline) to Missing (at post baseline)
1
10%
Sodium- CTCAE Graded Low: Grade 3 (at baseline) to Grade 3 (at post baseline)
1
10%
Sodium- CTCAE Graded High: Grade 0 (at baseline) to Grade 0 (at post baseline)
10
100%
11
100%
23
85.2%
22
81.5%
Sodium- CTCAE Graded High: Grade 0 (at baseline) to Grade 1 (at post baseline)
0
0%
0
0%
3
11.1%
5
18.5%
Sodium- CTCAE Graded High: Grade 0 (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
10. Secondary Outcome
Title Number of Participants With Shift From Baseline in Laboratory Parameter Values Based on Normal Range: Chemistry and Thyroid Function
Description Chemistry laboratory parameters: Blood urea nitrogen (BUN), Total protein, Chloride, Cancer antigen 19-9 (CA 19-9), Brain natriuretic peptide (BNP), Bicarbonate, Direct bilirubin, Carcinoembryonic antigen (CEA), Lactate dehydrogenase (LDH), Uric acid, Troponin I. Thyroid panel laboratory parameters: Thyroid-stimulating hormone (TSH), Free triiodothyronine (T3), Free thyroxine (T4). Laboratory values were as per laboratory normal ranges. Values above range were reported as high and values below range as low. Shift in chemistry and thyroid panel severity from baseline grade low, normal, high and missing to the post baseline grades as low, normal, high and missing are reported in this outcome measure. Categories with at least 1 non-zero data values are reported.
Time Frame Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
Safety set included of all participants who received at least 1 dose of study drug. Here "Number Analyzed" signifies number of participants evaluable for specified rows.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 27 27
BNP: Normal (at baseline) to Normal (at post baseline)
8
80%
6
54.5%
24
88.9%
22
81.5%
BNP: Normal (at baseline) to High (at post baseline)
1
10%
3
27.3%
2
7.4%
3
11.1%
BNP: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
BNP: High (at baseline) to Normal (at post baseline)
0
0%
2
18.2%
0
0%
BNP: High (at baseline) to High (at post baseline)
1
10%
0
0%
2
7.4%
BUN: Normal (at baseline) to Normal (at post baseline)
4
40%
3
27.3%
15
55.6%
14
51.9%
BUN (mmol/L): Normal (at baseline) to High (at post baseline)
3
30%
5
45.5%
9
33.3%
7
25.9%
BUN: Normal (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
1
3.7%
0
0%
BUN: High (at baseline) to Normal (at post baseline)
0
0%
0
0%
2
7.4%
3
11.1%
BUN: High (at baseline) to High (at post baseline)
3
30%
2
18.2%
0
0%
3
11.1%
Bicarbonate: Low (at baseline) to Normal (at post baseline)
1
10%
0
0%
3
11.1%
Bicarbonate: Low (at baseline) to High (at post baseline)
1
10%
1
9.1%
0
0%
Bicarbonate: Normal (at baseline) to Low (at post baseline)
2
20%
1
9.1%
6
22.2%
9
33.3%
Bicarbonate: Normal (at baseline) to Normal (at post baseline)
7
70%
6
54.5%
18
66.7%
11
40.7%
Bicarbonate: Normal (at baseline) to High (at post baseline)
1
10%
2
18.2%
1
3.7%
3
11.1%
Bicarbonate: Normal (at baseline) to High and Low (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
Bicarbonate: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
CA 19-9: Normal (at baseline) to Normal (at post baseline)
3
30%
3
27.3%
6
22.2%
7
25.9%
CA 19-9: Normal (at baseline) to High (at post baseline)
0
0%
0
0%
2
7.4%
1
3.7%
CA 19-9: Normal (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
0
0%
1
3.7%
CA 19-9: High (at baseline) to Normal (at post baseline)
0
0%
0
0%
1
3.7%
1
3.7%
CA 19-9: High (at baseline) to High (at post baseline)
7
70%
6
54.5%
16
59.3%
16
59.3%
CA 19-9: High (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
2
7.4%
1
3.7%
CEA: Normal (at baseline) to Normal (at post baseline)
1
10%
1
9.1%
3
11.1%
CEA: Normal (at baseline) to High (at post baseline)
0
0%
2
18.2%
2
7.4%
CEA: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
CEA: High (at baseline) to Normal (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
CEA: High (at baseline) to High (at post baseline)
9
90%
9
81.8%
20
74.1%
20
74.1%
CEA: High (at baseline) to Missing (at post baseline)
0
0%
2
18.2%
3
11.1%
1
3.7%
Chloride: Low (at baseline) to Low (at post baseline)
1
10%
1
9.1%
1
3.7%
Chloride: Low (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
0
0%
Chloride: Normal (at baseline) to Low (at post baseline)
2
20%
2
18.2%
8
29.6%
3
11.1%
Chloride: Normal (at baseline) to Normal (at post baseline)
8
80%
7
63.6%
15
55.6%
22
81.5%
Chloride: Normal (at baseline) to High (at post baseline)
0
0%
0
0%
2
7.4%
1
3.7%
Chloride: Normal (at baseline) to High & Low (at post baseline)
0
0%
1
9.1%
0
0%
0
0%
Free T3: Low (at baseline) to Low (at post baseline)
0
0%
1
9.1%
Free T3: Low (at baseline) to Missing (at post baseline)
1
10%
1
9.1%
Free T3: Normal (at baseline) to Low (at post baseline)
3
30%
5
45.5%
8
29.6%
7
25.9%
Free T3: Normal (at baseline) to Normal (at post baseline)
7
70%
5
45.5%
15
55.6%
15
55.6%
Free T3: Normal (at baseline) to High and Low (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Free T3: Normal (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
2
7.4%
1
3.7%
Free T3: High (at baseline) to High (at post baseline)
2
20%
Free T4: Low (at baseline) to Low (at post baseline)
0
0%
1
9.1%
Free T4: Low (at baseline) to Normal (at post baseline)
1
10%
0
0%
Free T4: Normal (at baseline) to Low (at post baseline)
0
0%
1
9.1%
2
7.4%
3
11.1%
Free T4: Normal (at baseline) to Normal (at post baseline)
10
100%
8
72.7%
17
63%
17
63%
Free T4: Normal (at baseline) to High (at post baseline)
0
0%
0
0%
3
11.1%
1
3.7%
Free T4: Normal (at baseline) to High and Low (at post baseline)
0
0%
0
0%
1
3.7%
1
3.7%
Free T4: Normal (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
3
11.1%
2
7.4%
Free T4: High (at baseline) to Low (at post baseline)
0
0%
1
9.1%
Free T4: High (at baseline) to Normal (at post baseline)
1
10%
1
9.1%
LDH: Low (at baseline) to Low (at post baseline)
1
10%
LDH: Normal (at baseline) to Normal (at post baseline)
1
10%
1
9.1%
6
22.2%
5
18.5%
LDH: Normal (at baseline) to High (at post baseline)
5
50%
4
36.4%
14
51.9%
10
37%
LDH: High (at baseline) to Normal (at post baseline)
0
0%
0
0%
0
0%
1
3.7%
LDH: High (at baseline) to High (at post baseline)
3
30%
6
54.5%
6
22.2%
11
40.7%
LDH: High (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Protein: Low (at baseline) to Low (at post baseline)
1
10%
Protein: Normal (at baseline) to Low (at post baseline)
5
50%
6
54.5%
11
40.7%
11
40.7%
Protein: Normal (at baseline) to Normal (at post baseline)
5
50%
4
36.4%
12
44.4%
14
51.9%
Protein: Normal (at baseline) to High (at post baseline)
0
0%
0
0%
0
0%
2
7.4%
Protein: Normal (at baseline) to High and Low (at post baseline)
0
0%
0
0%
3
11.1%
0
0%
Protein: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
TSH: Low (at baseline) to Low (at post baseline)
1
10%
TSH: Normal (at baseline) to Low (at post baseline)
0
0%
0
0%
2
7.4%
3
11.1%
TSH: Normal (at baseline) to Normal (at post baseline)
8
80%
7
63.6%
14
51.9%
20
74.1%
TSH: Normal (at baseline) to High (at post baseline)
1
10%
0
0%
3
11.1%
1
3.7%
TSH: Normal (at baseline) to High and Low (at post baseline)
0
0%
0
0%
2
7.4%
0
0%
TSH: Normal (at baseline) to Missing (at post baseline)
0
0%
0
0%
3
11.1%
2
7.4%
TSH: High (at baseline) to Normal (at post baseline)
0
0%
2
18.2%
2
7.4%
TSH: High (at baseline) to High (at post baseline)
1
10%
1
9.1%
1
3.7%
TSH: High (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
0
0%
Troponin I: Normal (at baseline) to Normal (at post baseline)
2
20%
11
100%
10
37%
Troponin I: Normal (at baseline) to High (at post baseline)
0
0%
1
9.1%
2
7.4%
Troponin I: Normal (at baseline) to Missing (at post baseline)
0
0%
14
127.3%
14
51.9%
Troponin I: Missing (at baseline) to Normal (at post baseline)
4
40%
3
27.3%
1
3.7%
Troponin I: Missing (at baseline) to High (at post baseline)
1
10%
0
0%
0
0%
Uric acid: Low (at baseline) to Low (at post baseline)
0
0%
0
0%
2
7.4%
Uric acid: Low (at baseline) to Normal (at post baseline)
1
10%
0
0%
1
3.7%
Uric acid: Low (at baseline) to Missing (at post baseline)
0
0%
1
9.1%
0
0%
Uric acid: Normal (at baseline) to Low (at post baseline)
1
10%
2
18.2%
4
14.8%
4
14.8%
Uric acid: Normal (at baseline) to Normal (at post baseline)
8
80%
5
45.5%
15
55.6%
16
59.3%
Uric acid: Normal (at baseline) to High (at post baseline)
0
0%
2
18.2%
4
14.8%
1
3.7%
Uric acid: High (at baseline) to Normal (at post baseline)
0
0%
0
0%
1
3.7%
0
0%
Uric acid: High (at baseline) to High (at post baseline)
1
10%
1
9.1%
2
7.4%
3
11.1%
11. Secondary Outcome
Title Number of Participants With Abnormal Hepatic Laboratory Values
Description Criteria for abnormal hepatic laboratory parameters: Aspartate aminotransferase (AST) and Alanine aminotransferase (ALT): >3* upper limit of normal (ULN), >5*ULN, >8*ULN, >10*ULN, >20*ULN; Total bilirubin (TBL) >1.5*ULN, >2*ULN; Alkaline phosphatase (ALP) >2*ULN, >3*ULN. Categories with at least 1 non-zero data values are reported.
Time Frame Phase 1b: Baseline up to 30 days after last dose (maximum up to 9 months approximately), Phase 2: Baseline up to 30 days after last dose (maximum up to 26 months approximately)

Outcome Measure Data

Analysis Population Description
Safety set included of all participants who received at least 1 dose of study drug. Here "Number Analyzed" signifies number of participants evaluable for specified rows.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 27 27
ALT: >3 ULN
0
0%
0
0%
2
7.4%
7
25.9%
ALT: >5 ULN
0
0%
0
0%
0
0%
5
18.5%
ALT: >8 ULN
0
0%
0
0%
0
0%
1
3.7%
AST: >3 ULN
0
0%
1
9.1%
4
14.8%
8
29.6%
AST: >5 ULN
0
0%
0
0%
1
3.7%
3
11.1%
AST: >8 ULN
0
0%
0
0%
1
3.7%
1
3.7%
ALT or AST: >3 ULN
0
0%
1
9.1%
3
11.1%
7
25.9%
ALT or AST: >5 ULN
0
0%
0
0%
1
3.7%
4
14.8%
ALT or AST: >8 ULN
0
0%
0
0%
1
3.7%
1
3.7%
Total bilirubin: >1.5 ULN
0
0%
2
18.2%
0
0%
1
3.7%
Alkaline phosphatase: >2 ULN
3
30%
3
27.3%
4
14.8%
8
29.6%
Alkaline phosphatase: >3 ULN
4
40%
4
36.4%
2
7.4%
4
14.8%
12. Secondary Outcome
Title Concentration Versus Time Summary of Plasma Concentration of Binimetinib
Description
Time Frame 1.5 hours post dose of binimetinib on Day 1, 15 of Cycle 1; pre dose of binimetinib on Day 15 of Cycle 1, 2, 3, 4, 5

Outcome Measure Data

Analysis Population Description
The pharmacokinetic analysis set included of all participants who received at least 1 dose of binimetinib and have at least one evaluable bioanalytical result. Here "Number Analyzed" signifies number of participants evaluable for specified rows.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
Measure Participants 10 11 26 27
Cycle 1 Day 1: 1.5 Hours post dose
228
(152.2)
163
(129.6)
261
(90.9)
217
(134.5)
Cycle 1 Day 15: pre dose
135
(60.9)
125
(103.9)
77.0
(141.2)
81.8
(143.6)
Cycle 1 Day 15: 1.5 Hours post dose
542
(57.6)
414
(24.5)
294
(84.4)
324
(83.3)
Cycle 2 Day 15: pre dose
108
(52.9)
NA
(NA)
88.5
(111.3)
120
(77.7)
Cycle 3 Day 15: pre dose
140
(42.0)
NA
(NA)
NA
(NA)
89.2
(183.4)
Cycle 4 Day 15: pre dose
NA
(NA)
NA
(NA)
NA
(NA)
NA
(NA)
Cycle 5 Day 15: pre dose
NA
(NA)
NA
(NA)
NA
(NA)

Adverse Events

Time Frame All-Cause Mortality: From start of the treatment up to 150 days after last dose (Phase 1b: maximum up to 13 months approximately and Phase 2: maximum up to 30 months approximately); TEAEs (including serious and other AEs): From start of the treatment up to 30 days after last dose (Phase 1b: maximum up to 9 months approximately and Phase 2: maximum up to 26 months approximately)
Adverse Event Reporting Description Same event may appear as both an adverse event (AE) and serious adverse event (SAE). However, what is presented are distinct events. An event may be categorized as serious in one participant and as non-serious in another, or a participant may have experienced both a serious and non-serious event. Safety set was evaluated. Both TEAE and treatment related AEs were monitored.
Arm/Group Title Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Arm/Group Description Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received binimetinib at a starting dose of 45 mg tablet BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion. Binimetinib dose modification to intermitted dosing (3 weeks on treatment and 1 week off treatment) with 45 mg dose or dose reduction to 30 mg BID or 30 mg intermittent dosing based on investigator's decision as per its tolerability among participants. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle, until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days. Participants with previously treated MSS metastatic colorectal cancer with RAS mutation received 45 mg binimetinib tablet orally BID along with 480 mg IV dose of nivolumab every 4 weeks in each 28 day treatment cycle. Ipilimumab was administered at the dose of 1 mg/kg IV every 8 weeks after completion of nivolumab infusion until disease progression, unacceptable toxicity, withdrawal of informal consent, initiation of subsequent anticancer therapy, lost to follow-up or death. Participants then followed up for safety for around 150 days.
All Cause Mortality
Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 8/10 (80%) 8/11 (72.7%) 20/27 (74.1%) 21/27 (77.8%)
Serious Adverse Events
Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 5/10 (50%) 6/11 (54.5%) 12/27 (44.4%) 11/27 (40.7%)
Cardiac disorders
Myocarditis 0/10 (0%) 1/11 (9.1%) 1/27 (3.7%) 0/27 (0%)
Pericardial effusion 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Acute coronary syndrome 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Cardiac failure 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Endocrine disorders
Hypophysitis 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Gastrointestinal disorders
Intestinal obstruction 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 1/27 (3.7%)
Abdominal pain 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Colitis 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Constipation 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Diarrhoea 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Ileus 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Nausea 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Pancreatitis 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Small intestinal obstruction 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Vomiting 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
General disorders
Pyrexia 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 1/27 (3.7%)
Asthenia 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Localised oedema 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Pain 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Hepatobiliary disorders
Hepatitis 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Infections and infestations
Pneumonia 1/10 (10%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Abdominal infection 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Bacterial sepsis 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Candida infection 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Cellulitis 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Empyema 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Infected seroma 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Klebsiella bacteraemia 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Lung infection 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Periorbital cellulitis 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Pneumocystis jirovecii pneumonia 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Pneumonia bacterial 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Injury, poisoning and procedural complications
Fall 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Hip fracture 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Infusion related reaction 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Investigations
Aspartate aminotransferase increased 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Blood creatine phosphokinase increased 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Transaminases increased 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Metabolism and nutrition disorders
Decreased appetite 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Hyponatraemia 0/10 (0%) 1/11 (9.1%) 1/27 (3.7%) 0/27 (0%)
Musculoskeletal and connective tissue disorders
Back pain 0/10 (0%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Myositis 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Pain in extremity 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Tumour associated fever 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Nervous system disorders
Ataxia 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Renal and urinary disorders
Nephritis 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Renal colic 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Respiratory, thoracic and mediastinal disorders
Pleural effusion 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Pneumonitis 0/10 (0%) 2/11 (18.2%) 0/27 (0%) 0/27 (0%)
Hypoxia 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Pleurisy 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Pulmonary oedema 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Skin and subcutaneous tissue disorders
Dermatitis acneiform 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Skin reaction 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Vascular disorders
Superior vena cava syndrome 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Other (Not Including Serious) Adverse Events
Phase 1b: Nivolumab+Binimetinib Phase 1b: Nivolumab+Ipilimumab+Binimetinib Phase 2: Nivolumab+Binimetinib Phase 2: Nivolumab+Ipilimumab+Binimetinib
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 10/10 (100%) 11/11 (100%) 27/27 (100%) 27/27 (100%)
Blood and lymphatic system disorders
Anaemia 0/10 (0%) 1/11 (9.1%) 7/27 (25.9%) 4/27 (14.8%)
Leukopenia 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Thrombocytopenia 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Cardiac disorders
Cardiac failure 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 1/27 (3.7%)
Ventricular hypokinesia 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Endocrine disorders
Hypothyroidism 1/10 (10%) 1/11 (9.1%) 0/27 (0%) 1/27 (3.7%)
Eye disorders
Visual impairment 2/10 (20%) 1/11 (9.1%) 0/27 (0%) 3/27 (11.1%)
Periorbital oedema 1/10 (10%) 0/11 (0%) 3/27 (11.1%) 1/27 (3.7%)
Vision blurred 1/10 (10%) 1/11 (9.1%) 1/27 (3.7%) 1/27 (3.7%)
Eyelid oedema 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Subretinal fluid 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 1/27 (3.7%)
Macular oedema 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Photopsia 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 1/27 (3.7%)
Retinopathy 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Dry age-related macular degeneration 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Eye oedema 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Vitreous detachment 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Gastrointestinal disorders
Diarrhoea 6/10 (60%) 4/11 (36.4%) 13/27 (48.1%) 15/27 (55.6%)
Nausea 5/10 (50%) 2/11 (18.2%) 12/27 (44.4%) 11/27 (40.7%)
Vomiting 2/10 (20%) 8/11 (72.7%) 7/27 (25.9%) 10/27 (37%)
Constipation 2/10 (20%) 3/11 (27.3%) 9/27 (33.3%) 6/27 (22.2%)
Abdominal pain 1/10 (10%) 0/11 (0%) 5/27 (18.5%) 5/27 (18.5%)
Dry mouth 0/10 (0%) 1/11 (9.1%) 3/27 (11.1%) 5/27 (18.5%)
Stomatitis 1/10 (10%) 2/11 (18.2%) 3/27 (11.1%) 3/27 (11.1%)
Abdominal distension 0/10 (0%) 1/11 (9.1%) 3/27 (11.1%) 3/27 (11.1%)
Dyspepsia 1/10 (10%) 1/11 (9.1%) 1/27 (3.7%) 3/27 (11.1%)
Gastrooesophageal reflux disease 1/10 (10%) 0/11 (0%) 0/27 (0%) 5/27 (18.5%)
Ascites 0/10 (0%) 0/11 (0%) 4/27 (14.8%) 0/27 (0%)
Abdominal discomfort 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 2/27 (7.4%)
Abdominal pain upper 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 2/27 (7.4%)
Abdominal pain lower 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Haemorrhoids 0/10 (0%) 1/11 (9.1%) 1/27 (3.7%) 0/27 (0%)
Pancreatitis 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Proctalgia 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Colitis 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Glossitis 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Lower gastrointestinal haemorrhage 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Mesenteric arterial occlusion 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Small intestinal obstruction 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
General disorders
Fatigue 4/10 (40%) 6/11 (54.5%) 11/27 (40.7%) 11/27 (40.7%)
Oedema peripheral 5/10 (50%) 5/11 (45.5%) 8/27 (29.6%) 11/27 (40.7%)
Pyrexia 3/10 (30%) 3/11 (27.3%) 8/27 (29.6%) 14/27 (51.9%)
Asthenia 3/10 (30%) 1/11 (9.1%) 7/27 (25.9%) 9/27 (33.3%)
Chills 1/10 (10%) 1/11 (9.1%) 4/27 (14.8%) 4/27 (14.8%)
Face oedema 0/10 (0%) 1/11 (9.1%) 4/27 (14.8%) 1/27 (3.7%)
Localised oedema 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 3/27 (11.1%)
Influenza like illness 0/10 (0%) 2/11 (18.2%) 1/27 (3.7%) 0/27 (0%)
Infections and infestations
Paronychia 1/10 (10%) 1/11 (9.1%) 3/27 (11.1%) 3/27 (11.1%)
Rash pustular 1/10 (10%) 1/11 (9.1%) 2/27 (7.4%) 3/27 (11.1%)
Urinary tract infection 0/10 (0%) 1/11 (9.1%) 2/27 (7.4%) 0/27 (0%)
Bronchitis 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 1/27 (3.7%)
Folliculitis 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Oral herpes 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 1/27 (3.7%)
Conjunctivitis 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Pneumonia 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Staphylococcal bacteraemia 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Injury, poisoning and procedural complications
Abdominal injury 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Fall 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 1/27 (3.7%)
Skin abrasion 1/10 (10%) 0/11 (0%) 0/27 (0%) 1/27 (3.7%)
Laceration 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Scratch 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Investigations
Blood creatine phosphokinase increased 5/10 (50%) 3/11 (27.3%) 14/27 (51.9%) 13/27 (48.1%)
Aspartate aminotransferase increased 0/10 (0%) 3/11 (27.3%) 2/27 (7.4%) 9/27 (33.3%)
Alanine aminotransferase increased 0/10 (0%) 2/11 (18.2%) 2/27 (7.4%) 8/27 (29.6%)
Ejection fraction decreased 1/10 (10%) 0/11 (0%) 4/27 (14.8%) 7/27 (25.9%)
Blood alkaline phosphatase increased 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 5/27 (18.5%)
Amylase increased 0/10 (0%) 0/11 (0%) 3/27 (11.1%) 3/27 (11.1%)
Blood bilirubin increased 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 2/27 (7.4%)
Lipase increased 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 2/27 (7.4%)
Gamma-glutamyltransferase increased 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Troponin T increased 0/10 (0%) 0/11 (0%) 0/27 (0%) 3/27 (11.1%)
Weight decreased 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Platelet count decreased 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Troponin I increased 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Electrocardiogram abnormal 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Intraocular pressure increased 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Prothrombin time prolonged 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Metabolism and nutrition disorders
Decreased appetite 2/10 (20%) 3/11 (27.3%) 13/27 (48.1%) 6/27 (22.2%)
Hypokalaemia 2/10 (20%) 1/11 (9.1%) 3/27 (11.1%) 2/27 (7.4%)
Hypomagnesaemia 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Dehydration 1/10 (10%) 1/11 (9.1%) 0/27 (0%) 1/27 (3.7%)
Hyperglycaemia 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Hypoalbuminaemia 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 1/27 (3.7%)
Hyponatraemia 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 2/27 (7.4%)
Hypophosphataemia 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 1/27 (3.7%)
Hypocalcaemia 0/10 (0%) 1/11 (9.1%) 1/27 (3.7%) 0/27 (0%)
Hyperuricaemia 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Musculoskeletal and connective tissue disorders
Back pain 0/10 (0%) 0/11 (0%) 5/27 (18.5%) 4/27 (14.8%)
Myalgia 0/10 (0%) 2/11 (18.2%) 3/27 (11.1%) 1/27 (3.7%)
Arthralgia 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Neck pain 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 2/27 (7.4%)
Muscular weakness 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 1/27 (3.7%)
Pain in extremity 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Musculoskeletal chest pain 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Tumour associated fever 1/10 (10%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Basal cell carcinoma 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Cancer pain 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Nervous system disorders
Dysgeusia 1/10 (10%) 2/11 (18.2%) 1/27 (3.7%) 2/27 (7.4%)
Dizziness 0/10 (0%) 2/11 (18.2%) 2/27 (7.4%) 0/27 (0%)
Neuropathy peripheral 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Headache 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Syncope 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Psychiatric disorders
Depression 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Confusional state 0/10 (0%) 2/11 (18.2%) 0/27 (0%) 0/27 (0%)
Insomnia 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Renal and urinary disorders
Haematuria 1/10 (10%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Proteinuria 0/10 (0%) 0/11 (0%) 1/27 (3.7%) 2/27 (7.4%)
Chromaturia 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Renal colic 0/10 (0%) 1/11 (9.1%) 1/27 (3.7%) 0/27 (0%)
Reproductive system and breast disorders
Benign prostatic hyperplasia 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Pruritus genital 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Scrotal oedema 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Respiratory, thoracic and mediastinal disorders
Cough 2/10 (20%) 3/11 (27.3%) 6/27 (22.2%) 6/27 (22.2%)
Dyspnoea 0/10 (0%) 4/11 (36.4%) 4/27 (14.8%) 4/27 (14.8%)
Pneumonitis 0/10 (0%) 2/11 (18.2%) 1/27 (3.7%) 1/27 (3.7%)
Dysphonia 0/10 (0%) 0/11 (0%) 3/27 (11.1%) 0/27 (0%)
Epistaxis 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 1/27 (3.7%)
Productive cough 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 2/27 (7.4%)
Rales 0/10 (0%) 2/11 (18.2%) 1/27 (3.7%) 0/27 (0%)
Wheezing 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Dyspnoea exertional 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Nasal congestion 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Skin and subcutaneous tissue disorders
Dermatitis acneiform 7/10 (70%) 6/11 (54.5%) 13/27 (48.1%) 12/27 (44.4%)
Rash 3/10 (30%) 1/11 (9.1%) 8/27 (29.6%) 14/27 (51.9%)
Pruritus 0/10 (0%) 3/11 (27.3%) 5/27 (18.5%) 7/27 (25.9%)
Dry skin 1/10 (10%) 2/11 (18.2%) 2/27 (7.4%) 5/27 (18.5%)
Rash maculo-papular 1/10 (10%) 1/11 (9.1%) 3/27 (11.1%) 0/27 (0%)
Alopecia 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 2/27 (7.4%)
Erythema 1/10 (10%) 1/11 (9.1%) 0/27 (0%) 2/27 (7.4%)
Pruritus generalised 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 2/27 (7.4%)
Rash macular 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 1/27 (3.7%)
Rash papular 0/10 (0%) 1/11 (9.1%) 1/27 (3.7%) 1/27 (3.7%)
Rash pruritic 2/10 (20%) 0/11 (0%) 1/27 (3.7%) 0/27 (0%)
Acne 0/10 (0%) 0/11 (0%) 0/27 (0%) 2/27 (7.4%)
Dermatitis 1/10 (10%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Hyperhidrosis 0/10 (0%) 0/11 (0%) 2/27 (7.4%) 0/27 (0%)
Dermatitis exfoliative generalised 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Ecchymosis 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Exfoliative rash 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Petechiae 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Rash erythematous 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Skin fissures 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Skin hyperpigmentation 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Skin lesion 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Skin reaction 0/10 (0%) 1/11 (9.1%) 0/27 (0%) 0/27 (0%)
Urticaria 1/10 (10%) 0/11 (0%) 0/27 (0%) 0/27 (0%)
Vascular disorders
Hypertension 0/10 (0%) 1/11 (9.1%) 3/27 (11.1%) 4/27 (14.8%)
Hypotension 1/10 (10%) 0/11 (0%) 1/27 (3.7%) 1/27 (3.7%)

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:
NCT03271047
Other Study ID Numbers:
  • ARRAY-162-202
  • C4211004
  • 2017-003464-12
First Posted:
Sep 1, 2017
Last Update Posted:
Jan 4, 2022
Last Verified:
Dec 1, 2021