An Open-Label, 2-Cohort, Multicenter, Study of Lenvatinib in Previously Treated Subjects With Unresectable Stage III or Stage IV Melanoma

Sponsor
Eisai Inc. (Industry)
Overall Status
Completed
CT.gov ID
NCT01136967
Collaborator
(none)
182
95
2
51
1.9
0

Study Details

Study Description

Brief Summary

The purpose of this study is to assess the objective response rate of lenvatinib in previously treated participants with American Joint Committee on Cancer (AJCC) unresectable Stage III or Stage IV melanoma and disease progression.

Condition or Disease Intervention/Treatment Phase
Phase 2

Detailed Description

This was a Phase 2, multicenter, open-label, 2-cohort, 2-stage study that assessed the ORR of lenvatinib in previously treated participants with AJCC unresectable Stage III or Stage IV melanoma and disease progression. Cohort 1 enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma, and is referred to as Cohort 1 or V600E BRAF negative. Other less common BRAF activating mutations were allowed as long as the participant did not receive a BRAF-targeted therapy. Cohort 2 enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy, and is referred to as Cohort 2 or V600E BRAF positive. Eligible participants had measurable disease according to RECIST 1.1.

Study Design

Study Type:
Interventional
Actual Enrollment :
182 participants
Allocation:
Non-Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
An Open-Label, 2-Cohort, Multicenter, Phase 2 Study of E7080 (Lenvatinib) in Previously Treated Subjects With Unresectable Stage III or Stage IV Melanoma
Actual Study Start Date :
Aug 1, 2010
Actual Primary Completion Date :
Apr 1, 2013
Actual Study Completion Date :
Nov 1, 2014

Arms and Interventions

Arm Intervention/Treatment
Experimental: Cohort 1 (V600E BRAF negative)

Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma.

Drug: Lenvatinib
Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles until disease progression, development of unacceptable toxicity or withdrawal of consent.
Other Names:
  • E7080
  • Experimental: Cohort 2 (V600E BRAF positive)

    Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy.

    Drug: Lenvatinib
    Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles until disease progression, development of unacceptable toxicity or withdrawal of consent.
    Other Names:
  • E7080
  • Outcome Measures

    Primary Outcome Measures

    1. Objective Response Rate (ORR) [From date of treatment start until all participants completed a minimum of 6 cycles (28-day cycles) or discontinued treatment prior to end of Cycle 6 (up to 24 weeks)]

      ORR, (ORR = CR + PR) was defined as the percentage of participants in each cohort who had a best overall response (BOR) of complete response (CR) or partial response (PR) based on Response Evaluation Criteria In Solid Tumors (RECIST) v1.1 for target lesions assessed by magnetic resonance imaging/computed tomography (MRI/CT) scans and independent radiologic review (IRR). A BOR of CR required confirmation by a subsequent CR assessment at least 4 weeks later. A BOR of PR required confirmation by a subsequent assessment of CR or PR at least 4 weeks later. CR was defined as the disappearance of all target lesions. Any pathological lymph nodes (target or non-target) had to have a reduction in short axis to less than (<)10 mm. PR was defined as at least a 30 percent (%) decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.

    Secondary Outcome Measures

    1. Progression Free Survival (PFS) [From date of treatment start until documentation of disease progression or death from any cause (whichever occurred first) or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months]

      PFS was measured as the time from the date of first administration of study treatment until the date of first documentation of disease progression or date of death from any cause (whichever occurred first), as determined by IRR and Investigator based on RECIST v1.1. Disease progression per RECIST v1.1 was defined as at least a 20% relative increase and 5 mm absolute increase in the sum of diameters of target lesions (taking as reference the smallest sum on study) recorded since the treatment started or the appearance of 1 or more new lesions. PFS was analyzed using Kaplan-Meier (1958) product-limit estimates. Data were presented with 2-sided 95% confidence interval (CI) when an adequate number of at risk participants warranted the estimates in the table below.

    2. Overall Survival (OS) [From date of treatment start until date of death from any cause or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months]

      OS was defined as the length of time in months from the date of first administration of study drug until the date of death from any cause, and was based on the data cutoff date for each cohort. OS was analyzed using Kaplan-Meier (1958) product-limit estimates. Data were presented with 2-sided 95% CI when an adequate number of at risk participants warranted the estimates in the table below.

    3. Disease Control Rate (DCR) [From date of treatment start until documentation of disease progression or death from any cause (whichever occurred first) or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months]

      DCR, (DCR = CR + PR + SD) was defined as the percentage of participants who had a BOR of CR or PR or stable disease (SD) based on RECIST v1.1 for target lesions assessed by MRI/CT and IRR. CR was defined as the disappearance of all target lesions, any pathological lymph nodes (target or non-target) had to have a reduction in short axis to <10 mm.; PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters; Overall Response (OR) = CR + PR. SD defined as reduction in tumor volume of < 30% or an increase in the volume of 1 or more measurable lesions of < 25% without the appearance of any new lesions which was neither tumor shrinkage corresponding to PR nor tumor expansion corresponding to disease progression. BOR of SD, time from first administration of study drug until date of documented SD needed to be >=7 weeks based on IRR and Investigator's assessment.

    4. Clinical Benefit Rate (CBR) [From date of treatment start until documentation of disease progression or death from any cause (whichever occurred first) or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months]

      CBR, (CBR = CR + PR + durable SD rate) was defined as the percentage of participants who had a BOR of CR or PR or durable SD (dSD, SD lasting >=23 weeks) based on RECIST v1.1 for target lesions assessed by MRI/CT, IRR and Investigator's assessment. A BOR of CR required confirmation by a subsequent CR assessment at least 4 weeks later. A BOR of PR required confirmation by a subsequent assessment of CR or PR at least 4 weeks later. CR was defined as the disappearance of all target lesions, any pathological lymph nodes (target or non-target) had to have a reduction in short axis to less than 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters; OR = CR + PR. A BOR of dSD, the time from the first administration of study drug until the date of documented dSD needed to be ≥23 weeks based on IRR and Investigator's assessment.

    5. Number of Participants With Adverse Events (AEs)/ Serious Adverse Events (SAEs) as a Measure of Safety and Tolerability of Lenvatinib [From date of treatment start up to 30 days after the last dose, or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 9 months]

      Safety was assessed by monitoring and recording all AEs including all Common Terminology Criteria for Adverse Events (CTCAE) grades and SAEs; regular monitoring of hematology, clinical chemistry, and urine values; physical examinations; and regular measurement of vital signs, electrocardiograms (ECGs), and multi-gated acquisition (MUGA) scans or echocardiogram.

    6. Change From Baseline in the Concentration of Clinical Biomarkers in Whole Blood [Cycle 1 Day 15 (C1 D15), Cycle 2 Day 1 (C2 D1), Cycle 3 Day 1 (C3 D1), Off-Treatment/Phase Visit 98 (V98)]

      Blood samples were drawn at specific time points. Utilizing a standard protocol, the deoxyribonucleic acid (DNA) from whole blood was extracted and analyzed for specific biomarkers of absorption, distribution, metabolism, and excretion of lenvatinib. Some of the biomarkers analyzed included; Angiopoietin, Epidermal Growth Factor (EGF), Fibroblast Growth Factor (FGF), FMS Like Tyrosine Kinase 3 Ligand (Flt3l) Granulocyte Colony Stimulating Factor (G-CSF), Granulocyte Macro Colony Stimulating Factor (GM-CSF), Interleukin 1 Receptor Antagonist (IL-1RA), Interferon (IFN), Macrophage Inflammatory Protein (MIP) 1 alpha, Platelet Derived Growth Factor (PDGF), Stromal Cell Derived Factor (SDF) 1 alpha, Interleukin (IL), Transforming Growth Factor (TGF), Tumor Necrosis Factor (TNF), Vascular Endothelial Growth Factor (VEGF).

    7. Summary of Plasma Concentration of Lenvatinib [Predose and 2 to 12 hours postdose at Cycle 1 Day 1 (C1D1), Cycle 1 Day 15 (C1D15), and Cycle 2 Day 1 (C2D1)]

      Blood samples for the quantification of lenvatinib in plasma were obtained and processed using a standardized protocol. The lower limit of quantification was 0.25 ng/mL. Pharmacokinetic (PK) analysis was conducted using nonlinear mixed effects modeling. Descriptive statistics were used to summarize lenvatinib plasma concentration data.

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    18 Years to 99 Years
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    1. Histologically confirmed diagnosis of melanoma.

    2. Unresectable Stage III or Stage IV melanoma.

    3. Evidence of disease progression according to RECIST 1.1 on prior regimen.

    4. Participants with brain metastases will be eligible if they have undergone complete surgical excision and are more then 1 month post surgery with no radiographic evidence of disease recurrence in the brain or have undergone stereotactic radio surgery (gamma knife procedure) and are more then 1 month post procedure and with no radiographic evidence of disease progression in the brain; and are asymptomatic, and discontinued corticosteroid treatment at least 30 days prior starting treatment.

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

    6. Adequately controlled blood pressure.

    7. Adequate renal function, bone marrow function, blood coagulation function, and liver function, as defined in the study protocol.

    Exclusion Criteria:
    1. Melanoma of intraocular origin.

    2. Leptomeningeal metastases or brain metastases except as for participants with brain metastases will be eligible if they have undergone complete surgical excision and are more then 1 month post surgery with no radiographic evidence of disease recurrence in the brain or have undergone stereotactic radio surgery (gamma knife procedure) and are more then 1 month post procedure and with no radiographic evidence of disease progression in the brain; and are asymptomatic, and discontinued corticosteroid treatment at least 30 days prior starting treatment.

    3. More than 2 prior systemic anticancer regimen treatments including immunotherapies for unresectable Stage III or Stage IV disease (if BRAF V600E mutation negative) or not previously treated with BRAF V600E-targeted therapy or received in the past more than 2 prior systemic anticancer regimen treatments, including immunotherapies, in addition to a BRAF-V600E-targeted therapy (if BRAF V600E mutation positive).

    4. Significant cardiovascular impairment.

    5. Bleeding disorder or a thrombotic disorder requiring anticoagulant therapy.

    6. Females who are pregnant or breastfeeding.

    7. Prolongation of QTc interval to greater than 480 msec.

    8. 24 hour urine protein greater than or equal to 1 gm.

    9. Active hemoptysis within 3 wks prior to the first dose of study drug.

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Birmingham Alabama United States
    2 Tucson Arizona United States
    3 Los Angeles California United States
    4 San Francisco California United States
    5 Aurora Colorado United States
    6 Boulder Colorado United States
    7 Colorado Springs Colorado United States
    8 Denver Colorado United States
    9 Lakewood Colorado United States
    10 Littleton Colorado United States
    11 Lone Tree Colorado United States
    12 Longmont Colorado United States
    13 Parker Colorado United States
    14 Thornton Colorado United States
    15 Bonita Springs Florida United States
    16 Bradenton Florida United States
    17 Cape Coral Florida United States
    18 Clearwater Florida United States
    19 Englewood Florida United States
    20 Fort Myers Florida United States
    21 Gainesville Florida United States
    22 Naples Florida United States
    23 North Port Florida United States
    24 Orlando Florida United States
    25 Palm Harbor Florida United States
    26 Port Charlotte Florida United States
    27 Sarasota Florida United States
    28 Sebring Florida United States
    29 Tampa Florida United States
    30 Venice Florida United States
    31 Louisville Kentucky United States
    32 Boston Massachusetts United States
    33 Detroit Michigan United States
    34 Burnsville Minnesota United States
    35 Coon Rapids Minnesota United States
    36 Edina Minnesota United States
    37 Fridley Minnesota United States
    38 Maplewood Minnesota United States
    39 Minneapolis Minnesota United States
    40 Saint Paul Minnesota United States
    41 Woodbury Minnesota United States
    42 Southaven Mississippi United States
    43 Saint Louis Missouri United States
    44 Henderson Nevada United States
    45 Las Vegas Nevada United States
    46 Lebanon New Hampshire United States
    47 Morristown New Jersey United States
    48 New York New York United States
    49 Cincinnati Ohio United States
    50 Cleveland Ohio United States
    51 Columbus Ohio United States
    52 Eugene Oregon United States
    53 Portland Oregon United States
    54 Springfield Oregon United States
    55 Tualatin Oregon United States
    56 Bethlehem Pennsylvania United States
    57 Philadelphia Pennsylvania United States
    58 Bartlett Tennessee United States
    59 Franklin Tennessee United States
    60 Gallatin Tennessee United States
    61 Germantown Tennessee United States
    62 Hermitage Tennessee United States
    63 Lebanon Tennessee United States
    64 Memphis Tennessee United States
    65 Murfreesboro Tennessee United States
    66 Nashville Tennessee United States
    67 Smyrna Tennessee United States
    68 Bedford Texas United States
    69 Dallas Texas United States
    70 Grapevine Texas United States
    71 Houston Texas United States
    72 Arlington Virginia United States
    73 Fairfax Virginia United States
    74 Gainesville Virginia United States
    75 Leesburg Virginia United States
    76 Winchester Virginia United States
    77 Woodbridge Virginia United States
    78 Vancouver Washington United States
    79 Madison Wisconsin United States
    80 Adelaide Australia
    81 Malvern Australia
    82 Newcastle Australia
    83 North Sydney Australia
    84 Perth Australia
    85 Westmead Australia
    86 Essen Germany
    87 Hannover Germany
    88 Heidelberg Germany
    89 Kiel Germany
    90 Mainz Germany
    91 Tubingen Germany
    92 Glasgow United Kingdom
    93 London United Kingdom
    94 Nottingham United Kingdom
    95 Surrey United Kingdom

    Sponsors and Collaborators

    • Eisai Inc.

    Investigators

    • Study Director: Eisai US Medical Services, Eisai Limited

    Study Documents (Full-Text)

    None provided.

    More Information

    Publications

    None provided.
    Responsible Party:
    Eisai Inc.
    ClinicalTrials.gov Identifier:
    NCT01136967
    Other Study ID Numbers:
    • E7080-G000-206
    First Posted:
    Jun 4, 2010
    Last Update Posted:
    Nov 13, 2019
    Last Verified:
    Nov 1, 2016
    Additional relevant MeSH terms:

    Study Results

    Participant Flow

    Recruitment Details A total of 298 participants were screened. Of these, 116 were screen failures and 182 received treatment (93 participants in Cohort 1 and 89 participants in Cohort 2).
    Pre-assignment Detail
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Period Title: Overall Study
    STARTED 93 89
    Disease Progression 55 56
    Treatment Ongoing at Data Cutoff 14 6
    COMPLETED 69 62
    NOT COMPLETED 24 27

    Baseline Characteristics

    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive) Total
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Total of all reporting groups
    Overall Participants 93 89 182
    Age (Years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [Years]
    62.5
    (11.71)
    55.0
    (14.18)
    58.8
    (13.47)
    Sex/Gender, Customized (Number) [Number]
    Female
    29
    31.2%
    39
    43.8%
    68
    37.4%
    Male
    64
    68.8%
    50
    56.2%
    114
    62.6%
    AJCC Melanoma Tumor, Node, Metastasis (TNM) Stage at Study Entry (Number) [Number]
    Unresectable Stage III
    5
    5.4%
    7
    7.9%
    12
    6.6%
    Unresectable Stage IV
    88
    94.6%
    82
    92.1%
    170
    93.4%

    Outcome Measures

    1. Primary Outcome
    Title Objective Response Rate (ORR)
    Description ORR, (ORR = CR + PR) was defined as the percentage of participants in each cohort who had a best overall response (BOR) of complete response (CR) or partial response (PR) based on Response Evaluation Criteria In Solid Tumors (RECIST) v1.1 for target lesions assessed by magnetic resonance imaging/computed tomography (MRI/CT) scans and independent radiologic review (IRR). A BOR of CR required confirmation by a subsequent CR assessment at least 4 weeks later. A BOR of PR required confirmation by a subsequent assessment of CR or PR at least 4 weeks later. CR was defined as the disappearance of all target lesions. Any pathological lymph nodes (target or non-target) had to have a reduction in short axis to less than (<)10 mm. PR was defined as at least a 30 percent (%) decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.
    Time Frame From date of treatment start until all participants completed a minimum of 6 cycles (28-day cycles) or discontinued treatment prior to end of Cycle 6 (up to 24 weeks)

    Outcome Measure Data

    Analysis Population Description
    Full Analysis Set (Intent-to-Treat [ITT] Analysis Set) included all participants who received at least 1 dose study drug.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    Number (95% Confidence Interval) [Percentage of participants]
    8.6
    9.2%
    9.0
    10.1%
    2. Secondary Outcome
    Title Progression Free Survival (PFS)
    Description PFS was measured as the time from the date of first administration of study treatment until the date of first documentation of disease progression or date of death from any cause (whichever occurred first), as determined by IRR and Investigator based on RECIST v1.1. Disease progression per RECIST v1.1 was defined as at least a 20% relative increase and 5 mm absolute increase in the sum of diameters of target lesions (taking as reference the smallest sum on study) recorded since the treatment started or the appearance of 1 or more new lesions. PFS was analyzed using Kaplan-Meier (1958) product-limit estimates. Data were presented with 2-sided 95% confidence interval (CI) when an adequate number of at risk participants warranted the estimates in the table below.
    Time Frame From date of treatment start until documentation of disease progression or death from any cause (whichever occurred first) or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months

    Outcome Measure Data

    Analysis Population Description
    Full Analysis Set (ITT Analysis Set) included all participants who received at least 1 dose study drug.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    Determined by IRR
    3.7
    1.8
    Determined by Investigator
    3.7
    2.3
    3. Secondary Outcome
    Title Overall Survival (OS)
    Description OS was defined as the length of time in months from the date of first administration of study drug until the date of death from any cause, and was based on the data cutoff date for each cohort. OS was analyzed using Kaplan-Meier (1958) product-limit estimates. Data were presented with 2-sided 95% CI when an adequate number of at risk participants warranted the estimates in the table below.
    Time Frame From date of treatment start until date of death from any cause or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months

    Outcome Measure Data

    Analysis Population Description
    Full Analysis Set (ITT Analysis Set) included all participants who received at least 1 dose study drug.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    Median (95% Confidence Interval) [Months]
    8.9
    6.3
    4. Secondary Outcome
    Title Disease Control Rate (DCR)
    Description DCR, (DCR = CR + PR + SD) was defined as the percentage of participants who had a BOR of CR or PR or stable disease (SD) based on RECIST v1.1 for target lesions assessed by MRI/CT and IRR. CR was defined as the disappearance of all target lesions, any pathological lymph nodes (target or non-target) had to have a reduction in short axis to <10 mm.; PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters; Overall Response (OR) = CR + PR. SD defined as reduction in tumor volume of < 30% or an increase in the volume of 1 or more measurable lesions of < 25% without the appearance of any new lesions which was neither tumor shrinkage corresponding to PR nor tumor expansion corresponding to disease progression. BOR of SD, time from first administration of study drug until date of documented SD needed to be >=7 weeks based on IRR and Investigator's assessment.
    Time Frame From date of treatment start until documentation of disease progression or death from any cause (whichever occurred first) or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months

    Outcome Measure Data

    Analysis Population Description
    Full Analysis Set (ITT Analysis Set) included all participants who received at least 1 dose study drug.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    Determined by IRR
    52.7
    56.7%
    34.8
    39.1%
    Determined by Investigator
    64.5
    69.4%
    48.3
    54.3%
    5. Secondary Outcome
    Title Clinical Benefit Rate (CBR)
    Description CBR, (CBR = CR + PR + durable SD rate) was defined as the percentage of participants who had a BOR of CR or PR or durable SD (dSD, SD lasting >=23 weeks) based on RECIST v1.1 for target lesions assessed by MRI/CT, IRR and Investigator's assessment. A BOR of CR required confirmation by a subsequent CR assessment at least 4 weeks later. A BOR of PR required confirmation by a subsequent assessment of CR or PR at least 4 weeks later. CR was defined as the disappearance of all target lesions, any pathological lymph nodes (target or non-target) had to have a reduction in short axis to less than 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters; OR = CR + PR. A BOR of dSD, the time from the first administration of study drug until the date of documented dSD needed to be ≥23 weeks based on IRR and Investigator's assessment.
    Time Frame From date of treatment start until documentation of disease progression or death from any cause (whichever occurred first) or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 8 months

    Outcome Measure Data

    Analysis Population Description
    Full Analysis Set (ITT Analysis Set) included all participants who received at least 1 dose study drug.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    Determined by IRR
    31.2
    33.5%
    14.6
    16.4%
    Determined by Investigator
    33.3
    35.8%
    20.2
    22.7%
    6. Secondary Outcome
    Title Number of Participants With Adverse Events (AEs)/ Serious Adverse Events (SAEs) as a Measure of Safety and Tolerability of Lenvatinib
    Description Safety was assessed by monitoring and recording all AEs including all Common Terminology Criteria for Adverse Events (CTCAE) grades and SAEs; regular monitoring of hematology, clinical chemistry, and urine values; physical examinations; and regular measurement of vital signs, electrocardiograms (ECGs), and multi-gated acquisition (MUGA) scans or echocardiogram.
    Time Frame From date of treatment start up to 30 days after the last dose, or up to data cutoff (Cohort 1; 15 Jan 2012 and Cohort 2; 15 Apr 2013), up to approximately 2 years 9 months

    Outcome Measure Data

    Analysis Population Description
    Safety Analysis Set included those participants who received at least 1 dose of study drug and had at least 1 post baseline safety evaluation.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    AEs
    93
    100%
    89
    100%
    SAEs
    39
    41.9%
    36
    40.4%
    7. Secondary Outcome
    Title Change From Baseline in the Concentration of Clinical Biomarkers in Whole Blood
    Description Blood samples were drawn at specific time points. Utilizing a standard protocol, the deoxyribonucleic acid (DNA) from whole blood was extracted and analyzed for specific biomarkers of absorption, distribution, metabolism, and excretion of lenvatinib. Some of the biomarkers analyzed included; Angiopoietin, Epidermal Growth Factor (EGF), Fibroblast Growth Factor (FGF), FMS Like Tyrosine Kinase 3 Ligand (Flt3l) Granulocyte Colony Stimulating Factor (G-CSF), Granulocyte Macro Colony Stimulating Factor (GM-CSF), Interleukin 1 Receptor Antagonist (IL-1RA), Interferon (IFN), Macrophage Inflammatory Protein (MIP) 1 alpha, Platelet Derived Growth Factor (PDGF), Stromal Cell Derived Factor (SDF) 1 alpha, Interleukin (IL), Transforming Growth Factor (TGF), Tumor Necrosis Factor (TNF), Vascular Endothelial Growth Factor (VEGF).
    Time Frame Cycle 1 Day 15 (C1 D15), Cycle 2 Day 1 (C2 D1), Cycle 3 Day 1 (C3 D1), Off-Treatment/Phase Visit 98 (V98)

    Outcome Measure Data

    Analysis Population Description
    The Safety Analysis set was used and included all participants who received at least one dose of lenvatinib and had at least 1 postbaseline safety evaluation. Number analyzed (n) signifies participants who were evaluable at specific time points for this outcome measure.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    Angiopoietin 2 76 C1D15
    -1015.69
    (946.596)
    Angiopoietin 2 76 C2D1
    -832.29
    (1257.287)
    Angiopoietin 2 76 C3D1
    -923.82
    (969.285)
    Angiopoietin 2 76 V98
    -947.00
    (825.037)
    Angiopoietin 2 90 C1D15
    -1225.322
    (1950.4525)
    Angiopoietin 2 90 C2D1
    -1100.235
    (1378.4621)
    Angiopoietin 1 C1D15
    -3033.030
    (8808.2059)
    -2717.533
    (16689.4536)
    Angiopoietin 1 C2D1
    -2802.306
    (10202.7814)
    -1743.569
    (15546.4804)
    Angiopoietin 1 C3D1
    -859.021
    (9994.6711)
    Angiopoietin 1 V98
    8107.400
    (17740.0516)
    Angiopoietin 2 C1D15
    -1462.552
    (1541.8074)
    -2154.269
    (3560.0029)
    Angiopoietin 2 C2D1
    -1225.462
    (1872.5841)
    -1884.439
    (2357.1979)
    Angiopoietin 2 C3D1
    -1255.485
    (1738.0891)
    Angiopoietin 2 V98
    -1374.183
    (1158.2897)
    CD40 Ligand C1D15
    -24020.778
    (62788.2464)
    -4678.661
    (41191.7303)
    CD40 Ligand C2D1
    -22957.613
    (73677.4378)
    -7254.754
    (37794.5541)
    CD40 Ligand C3D1
    -16700.900
    (48016.0710)
    CD40 Ligand V98
    2192.177
    (86909.9688)
    EGF 2 C1D15
    -46.493
    (101.0772)
    -0.516
    (219.9787)
    EGF 2 C2D1
    -39.636
    (125.0664)
    -29.803
    (121.1705)
    EGF 2 C3D1
    -37.600
    (113.8543)
    EGF 2 V98
    -2.230
    (191.4129)
    EGF 59 C1D15
    -37.07
    (81.051)
    EGF 59 C2D1
    -31.58
    (96.200)
    EGF 59 C3D1
    -24.49
    (93.102)
    EGF 59 V98
    10.17
    (137.755)
    EGF 80 C1D15
    -31.519
    (126.2579)
    EGF 80 C2D1
    -32.171
    (121.7805)
    Eotaxin-4 C1D15
    51.456
    (117.1266)
    46.538
    (128.398)
    Eotaxin-4 C2D1
    99.974
    (207.0243)
    60.919
    (114.6134)
    Eotaxin-4 C3D1
    89.878
    (108.6392)
    Eotaxin-4 V98
    104.613
    (157.7527)
    FGF 2 C1D15
    -4.294
    (51.3985)
    8.325
    (136.3433)
    FGF 2 C2D1
    -8.085
    (48.8273)
    -8.003
    (144.9054)
    FGF 2 C3D1
    7.443
    (45.7310)
    FGF 2 V98
    -3.917
    (62.3623)
    FGF 23 C1D15
    6.549
    (22.5297)
    FGF 23 C2D1
    6.496
    (27.2305)
    FGF 23 C3D1
    6.691
    (24.5203)
    FGF 23 V98
    13.430
    (36.7130)
    FGF 4 C1D15
    -24.226
    (100.6823)
    50.369
    (253.9626)
    FGF 4 C2D1
    -15.491
    (104.9991)
    55.785
    (264.4585)
    FGF 4 C3D1
    5.283
    (130.3452)
    FGF 4 V98
    33.860
    (185.4616)
    FGF Basic Form C1D15
    -0.597
    (18.6851)
    43.457
    (224.9856)
    FGF Basic Form C2D1
    -1.212
    (38.4131)
    64.237
    (328.6676)
    FGF Basic Form C3D1
    2.060
    (29.2405)
    FGF Basic Form V98
    18.384
    (28.6214)
    Flt3l C1D15
    -0.037
    (32.5371)
    -8.877
    (23.0368)
    Flt3l C2D1
    5.135
    (28.0359)
    -1.362
    (39.2251)
    Flt3l C3D1
    5.439
    (24.1985)
    Flt3l V98
    13.367
    (17.7279)
    Fractalkine C1D15
    -11.700
    (94.5115)
    38.670
    (237.5045)
    Fractalkine C2D1
    -1.175
    (70.0858)
    4.663
    (58.1950)
    Fractalkine C3D1
    -5.588
    (116.3955)
    Fractalkine V98
    15.838
    (90.6004)
    G-CSF C1D15
    4.329
    (20.8343)
    6.368
    (61.7181)
    G-CSF C2D1
    7.366
    (32.5552)
    12.601
    (72.5113)
    G-CSF C3D1
    11.292
    (24.1828)
    G-CSF V98
    34.653
    (101.7055)
    GM-CSF C1D15
    0.767
    (16.9314)
    44.665
    (220.9124)
    GM-CSF C2D1
    4.371
    (38.3355)
    139.491
    (725.0903)
    GM-CSF C3D1
    10.036
    (49.8538)
    GM-CSF V98
    -1.495
    (5.1831)
    Growth Regulated Oncogene C1D15
    -141.838
    (410.3579)
    Growth Regulated Oncogene C2D1
    -120.148
    (456.3073)
    Hepatocyte Growth Factor C1D15
    102.527
    (1309.4077)
    -91.790
    (781.2225)
    Hepatocyte Growth Factor C2D1
    -126.667
    (693.2257)
    -147.796
    (411.1836)
    Hepatocyte Growth Factor C3D1
    61.257
    (1062.3266)
    Hepatocyte Growth Factor V98
    404.700
    (456.8130)
    Interferon Gamma C1D15
    4.072
    (19.3994)
    33.743
    (100.3678)
    Interferon Gamma C2D1
    -0.392
    (14.8083)
    -10.164
    (170.9869)
    Interferon Gamma C3D1
    2.986
    (5.7591)
    Interferon Gamma V98
    2.455
    (5.2255)
    Interleukin 1 alpha C1D15
    0.159
    (7.9441)
    67.203
    (200.9806)
    Interleukin 1 alpha C2D1
    0.226
    (8.2728)
    -36.352
    (198.1416)
    Interleukin 1 alpha C3D1
    -0.140
    (13.7136)
    Interleukin 1 Beta C1D15
    0.279
    (1.9668)
    59.768
    (137.6657)
    Interleukin 1 Beta C2D1
    1.198
    (5.0044)
    27.690
    (61.2456)
    Interleukin 1 Beta C3D1
    1.882
    (11.1737)
    IL-1RA C1D15
    14.490
    (63.5869)
    46.381
    (132.3985)
    IL-1RA C2D1
    20.295
    (61.9799)
    38.131
    (142.7521)
    IL-1RA C3D1
    7.048
    (30.9383)
    IL-1RA V98
    84.614
    (178.1590)
    Interleukin 12 (p40) C1D15
    14.514
    (73.9806)
    5.458
    (28.8514)
    Interleukin 12 (p40) C2D1
    16.052
    (40.3849)
    918.03
    (3917.8689)
    Interleukin 12 (p40) C3D1
    11.183
    (36.3506)
    Interleukin 12 (p40) V98
    -8.130
    (5.7276)
    Interleukin 12 (p70) C1D15
    1.241
    (18.8429)
    27.919
    (62.8944)
    Interleukin 12 (p70) C2D1
    -5.622
    (28.6381)
    7.820
    (71.1505)
    Interleukin 12 (p70) C3D1
    -6.099
    (19.5015)
    Interleukin 12 (p70) V98
    11.275
    (20.0182)
    Interleukin 10 C1D15
    -5.170
    (38.0092)
    -7.611
    (22.1926)
    Interleukin 10 C2D1
    -1.901
    (33.6203)
    1.687
    (72.7629)
    Interleukin 10 C3D1
    4.136
    (10.8185)
    Interleukin 10 V98
    -28.065
    (41.7547)
    Interleukin 13 C1D15
    0.961
    (5.0066)
    8.574
    (13.4754)
    Interleukin 13 C2D1
    2.451
    (7.6374)
    8.536
    (18.5220)
    Interleukin 13 C3D1
    5.404
    (16.4719)
    Interleukin 15 C1D15
    0.128
    (2.2957)
    85.619
    (234.7106)
    Interleukin 15 C2D1
    1.403
    (4.1630)
    574.776
    (1709.9272)
    Interleukin 15 C3D1
    -0.178
    (3.6993)
    Interleukin 15 V98
    3.930
    (NA)
    Interleukin 17 C1D15
    3.434
    (8.6463)
    4.567
    (12.1008)
    Interleukin 17 C2D1
    1.264
    (8.0963)
    3.035
    (15.8736)
    Interleukin 17 C3D1
    0.011
    (3.4100)
    Interleukin 17 V98
    8.440
    (11.7663)
    Interleukin 2 C1D15
    -1.004
    (7.1024)
    173.878
    (397.2754)
    Interleukin 2 C2D1
    5.853
    (20.3294)
    191.410
    (461.1991)
    Interleukin 2 C3D1
    2.647
    (19.5979)
    Interleukin 4 C1D15
    8.169
    (30.0305)
    12.815
    (77.4310)
    Interleukin 4 C2D1
    20.167
    (77.0619)
    7.815
    (54.5735)
    Interleukin 4 C3D1
    6.583
    (20.3824)
    Interleukin 5 C1D15
    0.311
    (1.1314)
    6.088
    (7.5239)
    Interleukin 5 C2D1
    0.205
    (1.5931)
    11.382
    (9.8663)
    Interleukin 5 C3D1
    -0.327
    (1.1102)
    Interleukin 6 C1D15
    0.166
    (6.8774)
    3.784
    (30.0715)
    Interleukin 6 C2D1
    4.778
    (34.0254)
    5.524
    (32.1205)
    Interleukin 6 C3D1
    2.027
    (7.3121)
    Interleukin 6 V98
    3.160
    (6.6276)
    Interleukin 7 C1D15
    1.267
    (7.3732)
    2.167
    (8.8122)
    Interleukin 7 C2D1
    2.413
    (8.8367)
    5.744
    (18.4339)
    Interleukin 7 C3D1
    0.616
    (5.0590)
    Interleukin 7 V98
    20.760
    (28.0439)
    Interleukin 8 C1D15
    -5.801
    (46.1572)
    -20.769
    (52.1135)
    Interleukin 8 C2D1
    -5.431
    (43.4197)
    -14.303
    (57.9561)
    Interleukin 8 C3D1
    -7.348
    (43.0509)
    Interleukin 8 V98
    26.982
    (32.7795)
    IFN gamma Induced Protein 10 C1D15
    453.684
    (1446.5825)
    184.891
    (501.5782)
    IFN gamma Induced Protein 10 C2D1
    246.194
    (246.194)
    306.075
    (792.5621)
    IFN gamma Induced Protein 10 C3D1
    201.923
    (483.5088)
    IFN gamma Induced Protein 10 V98
    359.872
    (501.1082)
    Monocyte Chemotactic Protein 1 C1D15
    0.733
    (423.2721)
    -101.580
    (371.5381)
    Monocyte Chemotactic Protein 1 C2D1
    -19.036
    (372.5242)
    128.749
    (541.1034)
    Monocyte Chemotactic Protein 1 C3D1
    13.542
    (370.3700)
    Monocyte Chemotactic Protein 1 V98
    -128.627
    (1234.5888)
    MIP 1 alpha C1D15
    -1.842
    (11.9129)
    -0.320
    (38.5273)
    MIP 1 alpha C2D1
    -2.098
    (12.0121)
    -0.473
    (35.4447)
    MIP 1 alpha C3D1
    -1.167
    (15.9038)
    MIP 1 alpha V98
    7.563
    (7.8802)
    MIP 1 beta C1D15
    0.032
    (20.1490)
    5.746
    (64.1386)
    MIP 1 beta C2D1
    -1.198
    (31.4522)
    31.277
    (261.3460)
    MIP 1 beta C3D1
    1.850
    (15.208)
    MIP 1 beta V98
    7.518
    (11.9566)
    PDGF AA 31 P1 C1D15
    -183.36
    (2260.110)
    PDGF AA 31 P1 C2D1
    -268.83
    (2054.666)
    PDGF AB C1D15
    -36.637
    (353.6898)
    -121.278
    (325.1308)
    PDGF AB C2D1
    -99.852
    (338.5816)
    -80.778
    (363.1249)
    PDGF AB C3D1
    -7.702
    (311.6834)
    PDGF AB V98
    130.800
    (382.6282)
    PDGF BB C1D15
    -120.317
    (1758.7655)
    -567.664
    (1385.6740)
    PDGF BB C2D1
    -435.311
    (1961.7449)
    -371.888
    (1186.1)
    PDGF BB C3D1
    333.637
    (1516.1506)
    PDGF BB V98
    174.083
    (3221.9009)
    Placental Derived Growth Factor C1D15
    55.444
    (55.0671)
    55.0671
    (29.8457)
    Placental Derived Growth Factor C2D1
    50.439
    (58.2571)
    51.359
    (76.6390)
    Placental Derived Growth Factor C3D1
    61.080
    (58.2969)
    Placental Derived Growth Factor V98
    112.683
    (110.3416)
    Chemokine Ligand 5 C1D15
    -2373.94
    (46225.695)
    Chemokine Ligand 5 C2D1
    551.64
    (49492.450)
    SDF 1 alpha C1D15
    441.305
    (385.8988)
    506.768
    (487.7603)
    SDF 1 alpha C2D1
    520.051
    (510.8558)
    647.435
    (470.0962)
    SDF 1 alpha C3D1
    509.483
    (550.4383)
    SDF 1 alpha V98
    836.532
    (686.4434)
    Soluble IL2 Receptor alpha C1D15
    -25.869
    (86.0991)
    -414.669
    (765.1544)
    Soluble IL2 Receptor alpha C2D1
    -13.695
    (65.5643)
    -290.918
    (1023.3392)
    Soluble IL2 Receptor alpha C3D1
    -12.233
    (31.5415)
    Soluble IL2 Receptor alpha V98
    -19.238
    (43.0942)
    TGF alpha C1D15
    -0.874
    (4.6605)
    21.148
    (168.7766)
    TGF alpha C2D1
    -0.350
    (6.5375)
    -6.019
    (24.3258)
    TGF alpha C3D1
    1.165
    (11.8825)
    TGF alpha V98
    2.590
    (3.4672)
    Tie-2 C1D15
    -2971.948
    (2397.5234)
    -3573.856
    (2996.7139)
    Tie-2 C2D1
    -3447.692
    (3577.1791)
    -4088.214
    (3191.9812)
    Tie-2 C3D1
    -2811.765
    (3459.1934)
    Tie-2 V98
    -1924.000
    (3215.5684)
    TNF alpha C1D15
    -0.233
    (4.9747)
    1.451
    (24.8216)
    TNF alpha C2D1
    0.039
    (3.9540)
    1.741
    (15.0677)
    TNF alpha C3D1
    0.614
    (4.6336)
    TNF alpha V98
    3.027
    (4.7343)
    VEGF C1D15
    -13.294
    (112.6871)
    10.709
    (285.2533)
    VEGF C2D1
    -11.523
    (122.6064)
    -26.215
    (239.4960)
    VEGF C3D1
    16.732
    (160.3096)
    VEGF V98
    155.638
    (352.8309)
    VEGF A C1D15
    78.868
    (174.8011)
    92.748
    (273.6190)
    VEGF A C2D1
    76.041
    (194.1394)
    140.664
    (336.9135)
    VEGF A C3D1
    116.800
    (233.1657)
    VEGF A V98
    465.833
    (515.7676)
    VEGF D C1D15
    0.953
    (87.6513)
    3.404
    (42.7585)
    VEGF D C2D1
    -9.776
    (98.0905)
    11.043
    (116.5445)
    VEGF D C3D1
    9.894
    (57.1140)
    VEGF D V98
    -21.317
    (88.6689)
    VEGF Rec 1 C1D15
    593.613
    (3109.3343)
    -221.867
    (799.9202)
    VEGF Rec 1 C2D1
    -3.971
    (739.7875)
    -243.661
    (1031.2376)
    VEGF Rec 1 C3D1
    -209.863
    (809.7744)
    VEGF Rec 1 V98
    -15.567
    (90.6274)
    VEGF Rec 2 C1D15
    -10110.096
    (5626.0125)
    -8842.040
    (6078.7151)
    VEGF Rec 2 C2D1
    -9369.165
    (17982.0625)
    -10676.563
    (7064.0825)
    VEGF Rec 2 C3D1
    -11389.576
    (10955.1596)
    VEGF Rec 2 V98
    -9021.217
    (5921.9619)
    VEGF Rec 3 C1D15
    -1174.650
    (2226.7893)
    -2251.867
    (3231.1485)
    VEGF Rec 3 C2D1
    -1320.738
    (3138.3810)
    -2523.257
    (3550.7291)
    VEGF Rec 3 C3D1
    -1488.140
    (3687.8774)
    VEGF Rec 3 V98
    -1201.680
    (2441.5741)
    8. Secondary Outcome
    Title Summary of Plasma Concentration of Lenvatinib
    Description Blood samples for the quantification of lenvatinib in plasma were obtained and processed using a standardized protocol. The lower limit of quantification was 0.25 ng/mL. Pharmacokinetic (PK) analysis was conducted using nonlinear mixed effects modeling. Descriptive statistics were used to summarize lenvatinib plasma concentration data.
    Time Frame Predose and 2 to 12 hours postdose at Cycle 1 Day 1 (C1D1), Cycle 1 Day 15 (C1D15), and Cycle 2 Day 1 (C2D1)

    Outcome Measure Data

    Analysis Population Description
    The PK analysis set was used for analysis and included all participants who received at least one dose of lenvatinib and had at least one quantifiable lenvatinib concentration. Number analyzed (n) signifies participants who were evaluable at specific time points for this outcome measure.
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    Measure Participants 93 89
    C1D1 Pre-dose
    0
    (0)
    0
    (0)
    C1D1 Post-dose
    229.6
    (148.98)
    287.6
    (168.97)
    C1D15 Pre-dose
    56.8
    (82)
    71.9
    (104.09)
    C1D15 Post-dose
    284.0
    (141.71)
    332.1
    (221.98)
    C2D1 Pre-dose
    38.7
    (32.94)
    52.0
    (48.73)
    C2D1 Post-dose
    244.5
    (182.67)
    270.4
    (143.64)

    Adverse Events

    Time Frame From date of administration of first dose up to 30 days after the last dose, or up to data cutoff (15 Jan 2012 and 15 Apr 2013 for Cohort 1 and Cohort 2, respectively), up to approximately 33 months.
    Adverse Event Reporting Description
    Arm/Group Title Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Arm/Group Description Cohort 1 (V600E BRAF negative) enrolled participants not harboring the V600E BRAF mutation with disease progression following up to 2 prior systemic anticancer regimens (excluding anti-VEGF) for unresectable Stage III or Stage IV melanoma. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles. Cohort 2 (V600E BRAF positive) enrolled participants harboring the activating BRAF mutations (mainly the V600E mutation) with disease progression following BRAF V600E-targeted therapy. Participants received lenvatinib 24 mg orally, once daily continuously in 28-day cycles.
    All Cause Mortality
    Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total / (NaN) / (NaN)
    Serious Adverse Events
    Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 39/93 (41.9%) 36/89 (40.4%)
    Blood and lymphatic system disorders
    Anaemia 0/93 (0%) 1/89 (1.1%)
    Cardiac disorders
    Atrial thrombosis 1/93 (1.1%) 0/89 (0%)
    Cardiac failure congestive 1/93 (1.1%) 0/89 (0%)
    Supraventricular tachycardia 1/93 (1.1%) 0/89 (0%)
    Atrioventricular block second degree 0/93 (0%) 1/89 (1.1%)
    Myocardial infarction 0/93 (0%) 1/89 (1.1%)
    Pericardial effusion 0/93 (0%) 1/89 (1.1%)
    Endocrine disorders
    Hypothyroidism 0/93 (0%) 1/89 (1.1%)
    Gastrointestinal disorders
    Nausea 3/93 (3.2%) 3/89 (3.4%)
    Pancreatitis 2/93 (2.2%) 0/89 (0%)
    Vomiting 2/93 (2.2%) 1/89 (1.1%)
    Abdominal pain 1/93 (1.1%) 3/89 (3.4%)
    Abdominal pain upper 1/93 (1.1%) 0/89 (0%)
    Colitis 1/93 (1.1%) 0/89 (0%)
    Oesophageal spasm 1/93 (1.1%) 0/89 (0%)
    Rectal perforation 1/93 (1.1%) 0/89 (0%)
    Constipation 0/93 (0%) 1/89 (1.1%)
    Diarrhoea 0/93 (0%) 1/89 (1.1%)
    Enterocolitis 0/93 (0%) 1/89 (1.1%)
    Intestinal obstruction 0/93 (0%) 1/89 (1.1%)
    Intestinal perforation 0/93 (0%) 1/89 (1.1%)
    General disorders
    Pyrexia 1/93 (1.1%) 0/89 (0%)
    Fatigue 0/93 (0%) 2/89 (2.2%)
    General physical health deterioration 0/93 (0%) 2/89 (2.2%)
    Impaired healing 0/93 (0%) 1/89 (1.1%)
    Pain 0/93 (0%) 1/89 (1.1%)
    Hepatobiliary disorders
    Cholelithiasis 1/93 (1.1%) 0/89 (0%)
    Gallbladder perforation 1/93 (1.1%) 0/89 (0%)
    Cholecystitis 0/93 (0%) 1/89 (1.1%)
    Cholecystitis acute 0/93 (0%) 1/89 (1.1%)
    Hepatic failure 0/93 (0%) 1/89 (1.1%)
    Infections and infestations
    Pneumonia 2/93 (2.2%) 2/89 (2.2%)
    Appendicitis 1/93 (1.1%) 0/89 (0%)
    Catheter site infection 1/93 (1.1%) 0/89 (0%)
    Incision site infection 1/93 (1.1%) 0/89 (0%)
    Oral herpes 1/93 (1.1%) 0/89 (0%)
    Pelvic abscess 1/93 (1.1%) 0/89 (0%)
    Urinary tract infection 1/93 (1.1%) 0/89 (0%)
    Cellulitis 0/93 (0%) 1/89 (1.1%)
    Infection 0/93 (0%) 1/89 (1.1%)
    Peritonitis 0/93 (0%) 1/89 (1.1%)
    Sepsis 0/93 (0%) 1/89 (1.1%)
    Septic shock 0/93 (0%) 1/89 (1.1%)
    Upper respiratory tract infection 0/93 (0%) 1/89 (1.1%)
    Wound infection 0/93 (0%) 1/89 (1.1%)
    Injury, poisoning and procedural complications
    Chemical peritonitis 1/93 (1.1%) 0/89 (0%)
    Spinal compression fracture 0/93 (0%) 1/89 (1.1%)
    Investigations
    Ejection fraction decreased 1/93 (1.1%) 0/89 (0%)
    Lipase increased 1/93 (1.1%) 0/89 (0%)
    Weight decreased 0/93 (0%) 1/89 (1.1%)
    Metabolism and nutrition disorders
    Hyponatraemia 2/93 (2.2%) 0/89 (0%)
    Dehydration 1/93 (1.1%) 1/89 (1.1%)
    Failure to thrive 0/93 (0%) 1/89 (1.1%)
    Hyperuricaemia 0/93 (0%) 1/89 (1.1%)
    Musculoskeletal and connective tissue disorders
    Back pain 1/93 (1.1%) 0/89 (0%)
    Myopathy 1/93 (1.1%) 0/89 (0%)
    Pain in extremity 1/93 (1.1%) 0/89 (0%)
    Groin pain 0/93 (0%) 2/89 (2.2%)
    Musculoskeletal chest pain 0/93 (0%) 1/89 (1.1%)
    Musculoskeletal pain 0/93 (0%) 1/89 (1.1%)
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Infected neoplasm 1/93 (1.1%) 0/89 (0%)
    Squamous cell carcinoma of skin 1/93 (1.1%) 0/89 (0%)
    Tumour pain 1/93 (1.1%) 1/89 (1.1%)
    Metastases to central nervous system 0/93 (0%) 1/89 (1.1%)
    Metastatic pain 0/93 (0%) 1/89 (1.1%)
    Nervous system disorders
    Cerebrovascular accident 2/93 (2.2%) 0/89 (0%)
    Hypoaesthesia 1/93 (1.1%) 0/89 (0%)
    Somnolence 1/93 (1.1%) 0/89 (0%)
    Syncope 1/93 (1.1%) 0/89 (0%)
    Cranial nerve palsies multiple 0/93 (0%) 1/89 (1.1%)
    Haemorrhage intracranial 0/93 (0%) 1/89 (1.1%)
    Headache 0/93 (0%) 1/89 (1.1%)
    Presyncope 0/93 (0%) 1/89 (1.1%)
    Spinal cord compression 0/93 (0%) 1/89 (1.1%)
    Psychiatric disorders
    Mental status changes 2/93 (2.2%) 1/89 (1.1%)
    Confusional state 1/93 (1.1%) 0/89 (0%)
    Renal and urinary disorders
    Renal failure 1/93 (1.1%) 0/89 (0%)
    Renal failure acute 1/93 (1.1%) 0/89 (0%)
    Urethral obstruction 1/93 (1.1%) 0/89 (0%)
    Urinary retention 1/93 (1.1%) 0/89 (0%)
    Respiratory, thoracic and mediastinal disorders
    Pulmonary embolism 3/93 (3.2%) 2/89 (2.2%)
    Dyspnoea 0/93 (0%) 1/89 (1.1%)
    Pleural effusion 0/93 (0%) 1/89 (1.1%)
    Skin and subcutaneous tissue disorders
    Angioedema 1/93 (1.1%) 0/89 (0%)
    Dermatitis bullous 1/93 (1.1%) 0/89 (0%)
    Vascular disorders
    Hypertension 5/93 (5.4%) 1/89 (1.1%)
    Deep vein thrombosis 3/93 (3.2%) 0/89 (0%)
    Aortic aneurysm 1/93 (1.1%) 0/89 (0%)
    Hypertensive crisis 1/93 (1.1%) 1/89 (1.1%)
    Hypotension 1/93 (1.1%) 0/89 (0%)
    Other (Not Including Serious) Adverse Events
    Cohort 1 (V600E BRAF Negative) Cohort 2 (V600E BRAF Positive)
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 93/93 (100%) 88/89 (98.9%)
    Blood and lymphatic system disorders
    Anaemia 5/93 (5.4%) 6/89 (6.7%)
    Thrombocytopenia 8/93 (8.6%) 9/89 (10.1%)
    Endocrine disorders
    Hypothyroidism 17/93 (18.3%) 24/89 (27%)
    Gastrointestinal disorders
    Abdominal pain 21/93 (22.6%) 19/89 (21.3%)
    Abdominal pain upper 11/93 (11.8%) 6/89 (6.7%)
    Constipation 25/93 (26.9%) 28/89 (31.5%)
    Diarrhoea 44/93 (47.3%) 26/89 (29.2%)
    Dry mouth 14/93 (15.1%) 10/89 (11.2%)
    Dyspepsia 5/93 (5.4%) 9/89 (10.1%)
    Flatulence 12/93 (12.9%) 2/89 (2.2%)
    Gastrooesophageal reflux disease 10/93 (10.8%) 6/89 (6.7%)
    Glossodynia 6/93 (6.5%) 2/89 (2.2%)
    Nausea 48/93 (51.6%) 36/89 (40.4%)
    Oral pain 10/93 (10.8%) 5/89 (5.6%)
    Stomatitis 16/93 (17.2%) 12/89 (13.5%)
    Vomiting 36/93 (38.7%) 32/89 (36%)
    General disorders
    Asthenia 6/93 (6.5%) 5/89 (5.6%)
    Chills 6/93 (6.5%) 4/89 (4.5%)
    Fatigue 62/93 (66.7%) 41/89 (46.1%)
    Oedema peripheral 14/93 (15.1%) 10/89 (11.2%)
    Pain 6/93 (6.5%) 5/89 (5.6%)
    Pyrexia 7/93 (7.5%) 10/89 (11.2%)
    Infections and infestations
    Oral herpes 5/93 (5.4%) 0/89 (0%)
    Upper respiratory tract infection 5/93 (5.4%) 4/89 (4.5%)
    Urinary tract infection 10/93 (10.8%) 9/89 (10.1%)
    Investigations
    Aspartate aminotransferase increased 3/93 (3.2%) 5/89 (5.6%)
    Blood alkaline phosphatase increased 7/93 (7.5%) 3/89 (3.4%)
    Blood thyroid stimulating hormone increased 14/93 (15.1%) 7/89 (7.9%)
    Gamma-glutamyltransferase increased 5/93 (5.4%) 1/89 (1.1%)
    Lipase increased 7/93 (7.5%) 3/89 (3.4%)
    Weight decreased 22/93 (23.7%) 12/89 (13.5%)
    Metabolism and nutrition disorders
    Decreased appetite 40/93 (43%) 30/89 (33.7%)
    Dehydration 6/93 (6.5%) 12/89 (13.5%)
    Hypokalaemia 6/93 (6.5%) 2/89 (2.2%)
    Hyponatraemia 8/93 (8.6%) 4/89 (4.5%)
    Musculoskeletal and connective tissue disorders
    Arthralgia 29/93 (31.2%) 20/89 (22.5%)
    Back pain 17/93 (18.3%) 12/89 (13.5%)
    Muscle spasms 7/93 (7.5%) 5/89 (5.6%)
    Muscular weakness 8/93 (8.6%) 3/89 (3.4%)
    Musculoskeletal chest pain 3/93 (3.2%) 6/89 (6.7%)
    Musculoskeletal pain 6/93 (6.5%) 6/89 (6.7%)
    Musculoskeletal stiffness 7/93 (7.5%) 4/89 (4.5%)
    Myalgia 13/93 (14%) 9/89 (10.1%)
    Pain in extremity 16/93 (17.2%) 9/89 (10.1%)
    Nervous system disorders
    Dizziness 10/93 (10.8%) 16/89 (18%)
    Dysgeusia 12/93 (12.9%) 16/89 (18%)
    Headache 29/93 (31.2%) 21/89 (23.6%)
    Lethargy 1/93 (1.1%) 5/89 (5.6%)
    Peripheral sensory neuropathy 10/93 (10.8%) 2/89 (2.2%)
    Psychiatric disorders
    Confusional state 2/93 (2.2%) 5/89 (5.6%)
    Depression 4/93 (4.3%) 5/89 (5.6%)
    Insomnia 7/93 (7.5%) 8/89 (9%)
    Renal and urinary disorders
    Haematuria 5/93 (5.4%) 3/89 (3.4%)
    Proteinuria 23/93 (24.7%) 17/89 (19.1%)
    Respiratory, thoracic and mediastinal disorders
    Cough 18/93 (19.4%) 11/89 (12.4%)
    Dysphonia 26/93 (28%) 33/89 (37.1%)
    Dyspnoea 14/93 (15.1%) 13/89 (14.6%)
    Epistaxis 8/93 (8.6%) 8/89 (9%)
    Oropharyngeal pain 13/93 (14%) 5/89 (5.6%)
    Skin and subcutaneous tissue disorders
    Dry skin 16/93 (17.2%) 7/89 (7.9%)
    Pruritus 7/93 (7.5%) 5/89 (5.6%)
    Rash 16/93 (17.2%) 5/89 (5.6%)
    Vascular disorders
    Hot flush 5/93 (5.4%) 1/89 (1.1%)
    Hypertension 53/93 (57%) 48/89 (53.9%)
    Hypotension 4/93 (4.3%) 6/89 (6.7%)

    Limitations/Caveats

    [Not Specified]

    More Information

    Certain Agreements

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

    Results Point of Contact

    Name/Title Eisai Medical Services
    Organization Eisai, Inc.
    Phone 1-888-422-4743
    Email esi_medinfo@eisai.com
    Responsible Party:
    Eisai Inc.
    ClinicalTrials.gov Identifier:
    NCT01136967
    Other Study ID Numbers:
    • E7080-G000-206
    First Posted:
    Jun 4, 2010
    Last Update Posted:
    Nov 13, 2019
    Last Verified:
    Nov 1, 2016