search
Back to results

A Study of Mobocertinib in Japanese Adults With Non-Small Cell Lung Cancer

Primary Purpose

Non-Small Cell Lung Cancer

Status
Active
Phase
Phase 1
Locations
Japan
Study Type
Interventional
Intervention
Mobocertinib
Sponsored by
Takeda
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Non-Small Cell Lung Cancer

Eligibility Criteria

20 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

General Inclusion Criteria (Both in Phase 1 and Phase 2 Part);

  1. Male or female patients ≥20 years old.
  2. Must have measurable disease by RECIST v1.1. Previously irradiated lesions may not be used for target lesions, unless there is unambiguous radiological progression after radiotherapy.
  3. Eastern Cooperative Oncology Group (ECOG) performance status 0 to 1.
  4. Minimum life expectancy of 3 months or more.
  5. Adequate renal and hepatic function as defined by the following criteria:

    •Total serum bilirubin ≤1.5 × upper limit of normal (ULN) (≤3.0 × ULN for patients with Gilbert syndrome or if liver function abnormalities are due to underlying malignancy);

    •Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) ≤2.5 × ULN (or ≤5 × ULN if liver function abnormalities are due to underlying malignancy);

    •Estimated creatinine clearance ≥30 mL/min (calculated by using the Cockcroft-Gault equation);

    •Serum albumin ≥2 g/dL; and

    •Serum lipase ≤1.5 × ULN; and

    •Serum amylase ≤1.5 × ULN unless the increased serum amylase is due to salivary isoenzymes.

  6. Adequate bone marrow function as defined by the following criteria:

    • Absolute neutrophil count ≥1.5 × 109/L;
    • Platelet count ≥75 × 109/L in Phase 1 Part and ≥100 × 109/L in Phase 2 Part; and
    • Hemoglobin ≥9.0 g/dL.
  7. Normal QT interval on screening ECG, defined as QTcF of ≤450 ms in males or ≤470 ms in females.
  8. Female patients who:

    • Are postmenopausal (natural amenorrhea and not due to other medical reasons) for at least 1 year before the screening visit, OR
    • Are surgically sterile, OR
    • If they are of childbearing potential, agree to practice 1 highly effective non-hormonal method of contraception and 1 additional effective (barrier) method (see Section 8.7.1) at the same time, from the time of signing the informed consent form (ICF) through 30 days after the last dose of study drug, OR
    • Agree to practice true abstinence, when this is in line with the preferred and usual lifestyle of the subject.

Note: Periodic abstinence (eg, calendar, ovulation, symptothermal, postovulation methods), withdrawal, spermicides only, and lactational amenorrhea are not acceptable methods of contraception.

Male patients, even if surgically sterilized (ie, status postvasectomy), who:

  • Agree to practice effective barrier contraception during the entire study treatment period and through 30 days after the last dose of study drug, OR
  • Agree to practice true abstinence, when this is in line with the preferred and usual lifestyle of the subject.

Note: Periodic abstinence (eg, calendar, ovulation, symptothermal, postovulation methods), withdrawal, spermicides only, and lactational amenorrhea are not acceptable methods of contraception.

9.Voluntary written consent must be given before performance of any study related procedure not part of standard medical care, with the understanding that consent may be withdrawn by the patient at any time without prejudice to future medical care.

10.Willingness and ability to comply with scheduled visits and study procedures.

  • Phase-Specific Inclusion Criteria (Phase 1 part);

    1.Have histologically or cytologically confirmed locally advanced (and not a candidate for definitive therapy) (Stage IIIB) or metastatic NSCLC (Stage IV).

    2.Refractory to standard available therapies. 3.All toxicities from prior therapy have resolved to ≤ grade 1 according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE v5.0), or have resolved to baseline, at the time of first dose of TAK-788. Note: treatment-related grade >1 alopecia or treatment-related grade 2 peripheral neuropathy are allowed if deemed irreversible.

  • Phase-Specific Inclusion Criteria (Phase 2 part);

    1. Histologically or cytologically confirmed locally advanced not suitable for definitive therapy, recurrent, or metastatic (Stage IV) NSCLC.
    2. Not received prior systemic treatment for locally advanced or metastatic disease (with the exception below): Neoadjuvant or adjuvant chemotherapy/immunotherapy for Stage I to III or combined modality chemotherapy/radiation for locally advanced disease is allowed if completed >6 months before the development of metastatic disease.
    3. A documented EGFR in-frame exon 20 insertion (including A763_Y764insFQEA, V769_D770insASV, D770_N771insNPG, D770_N771insSVD, H773_V774insNPH, or any other in-frame exon 20 insertion mutation) by a local test that has been analytically validated per local authority guidelines. The EGFR exon 20 insertion mutation can be either alone or in combination with other EGFR or HER2 mutations except EGFR common mutations (exon 19 del or L858R).
    4. Adequate tumor tissue available, either from primary or metastatic sites, for central laboratory confirmation of EGFR in-frame exon 20 insertion mutation. Note: confirmation of central test positivity is not required before the first dose of TAK-788.

      Exclusion Criteria:

      General Exclusion Criteria (Both in Phase 1 and Phase 2 Part);

    1. Have been diagnosed with another primary malignancy other than NSCLC except for adequately treated non-melanoma skin cancer or cervical cancer in situ; definitively treated non-metastatic prostate cancer; or patients with another primary malignancy who are definitively relapse-free with at least 3 years elapsed since the diagnosis of the other primary malignancy.
    2. Have undergone major surgery within 28 days prior to first dose of TAK-788. Minor surgical procedures, such as catheter placement or minimally invasive biopsy, are allowed.
    3. Have current spinal cord compression (symptomatic or asymptomatic and detected by radiographic imaging) or leptomeningeal disease (symptomatic or asymptomatic).
    4. Have significant, uncontrolled, or active cardiovascular disease, including, but not restricted to:
  • Myocardial infarction within 6 months prior to the first dose of study drug;
  • Unstable angina within 6 months prior to first dose;
  • Congestive heart failure within 6 months prior to first dose;
  • History of clinically significant (as determined by the treating physician) atrial arrhythmia;
  • Any history of ventricular arrhythmia; or
  • Cerebrovascular accident or transient ischemic attack within 6 months prior to first dose.

    5.Have uncontrolled hypertension. Patients with hypertension should be under treatment on study entry to control blood pressure.

    6.Currently being treated with medications known to be associated with the development of Torsades de Pointes.

    7.Have an ongoing or active infection, including, but not limited to, the requirement for intravenous antibiotics. Have a known history of HIV infection. Testing of HIV is not required in the absence of history.Hepatitis B surface antigen (HBsAg) positive patients are allowed to enroll if hepatitis B virus (HBV)-DNA is below 1000 copies/mL in the plasma.Patients who have positive hepatitis C virus (HCV) antibody can be enrolled but must have HCV-RNA undetectable in the plasma.

    8.Currently have or have a history of interstitial lung disease (ILD), radiation pneumonitis that required steroid treatment, or drug-related pneumonitis.

    9.Female patients who are lactating and breastfeeding or have a positive serum pregnancy test during the screening period.

  • Note: Female patients who are lactating will be eligible if they discontinue breastfeeding.

    10.Have gastrointestinal illness or disorder that could affect oral absorption of TAK-788.

    11.Have any condition or illness that, in the opinion of the investigator, might compromise patient safety or interfere with the evaluation of the safety of the drug.

  • Phase-Specific Exclusion Criteria (Phase 1 part);

    1. Previously received TAK-788.
    2. Received small-molecule anticancer therapy (including cytotoxic chemotherapy and investigational agents) within 14 days prior to the first dose of TAK-788 (except for reversible EGFR TKIs [ie, erlotinib or gefitinib] up to 7 days prior to the first dose of TAK-788).
    3. Received antineoplastic monoclonal antibodies including immunotherapy within 28 days prior to the first dose of TAK-788.
    4. Received radiotherapy within 14 days prior to the first dose of TAK-788, Stereotactic radiosurgery (SRS) and stereotactic body radiosurgery are allowed up to 7 days prior to the first dose.
    5. Have symptomatic CNS metastases (parenchymal or leptomeningeal) at screening or asymptomatic disease requiring corticosteroids to control symptoms within 7 days prior to the first dose of TAK-788.

      Note: If a patient has worsening neurological symptoms or signs due to CNS metastases, the patient needs to complete local therapy and be neurologically stable (with no requirement for corticosteroids or use of anticonvulsants) for 7 days prior to the first dose of TAK-788. Patients with no prior history of signs or symptoms of CNS metastases but who receive prophylactic steroids or anticonvulsants are allowed.

    6. Received a strong cytochrome P450 (CYP)3A inhibitor or strong CYP3A inducer within 2 weeks prior to first dose of TAK-788.
  • Phase-Specific Exclusion Criteria (Phase 2 part);

    1. Received radiotherapy within 14 days before the first dose of TAK-788 or has not recovered from radiotherapy-related toxicities. Stereotactic radiosurgery, stereotactic body radiotherapy, or palliative radiation outside the chest and brain is allowed up to 7 days before the first dose of TAK-788.
    2. Have known active brain metastases (have either previously untreated intracranial CNS metastases or previously treated intracranial CNS metastases with radiologically documented new or progressing CNS lesions). Brain metastases are allowed if they have been treated with surgery and/or radiation and have been stable without requiring corticosteroids to control symptoms within 7 days before the first dose of TAK-788, and have no evidence of new or enlarging brain metastases.
    3. Received a moderate or strong CYP3A inhibitor or moderate or strong CYP3A inducer within 10 days prior to first dose of TAK-788.
    4. Have cardiac ejection fraction <50% by echocardiogram or multigated acquisition (MUGA) scan at screening.

Sites / Locations

  • Aichi Cancer Center Hospital
  • Fujita Health University Hospital
  • National Cancer Center Hospital East
  • Kurume University Hospital
  • Hokkaido Cancer Center
  • Hyogo Cancer Cente
  • Kanazawa University Hospital
  • Kanagawa Cancer Center
  • Sendai Kousei Hospital
  • Okayama University Hospital
  • Kansai Medical University Hospital
  • Kindai University Hospital
  • National Hospital Organization Kinki-Chuo Chest Medical Center
  • Saitama Cancer Center
  • Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital
  • National Cancer Center Hospital
  • Cancer Institute Hospital
  • Yamaguchi Ube Medical Center
  • Kyushu University Hospital
  • Hiroshima University Hospital
  • Kyoto University Hospital
  • Niigata Cancer Center Hospital
  • Osaka International Cancer Institute
  • Tokushima University Hospital
  • Wakayama Medical University Hospital

Arms of the Study

Arm 1

Arm 2

Arm Type

Experimental

Experimental

Arm Label

Mobocertinib, Phase 1 Part

Mobocertinib, Phase 2 Part

Arm Description

Mobocertinib 40 milligrams (mg) (as the starting dose), capsules, orally, once daily on Days 1-28 of each 28-day treatment cycle for up to disease progression or intolerable toxicity, or another discontinuation criterion, and increasing until 160 mg, once daily (for up to approximately 10-12 cycles).

Mobocertinib 160 mg, once daily, for up to approximately 10-12 cycles.

Outcomes

Primary Outcome Measures

Phase 1 Part: Recommended Phase 2 Dose (RP2D) of Orally Administered Mobocertinib
The RP2D was the maximum tolerated dose (MTD) or less. The MTD was declared when at least 9 participants were evaluable in the study and 6 participants were evaluable at the current dose, and the current dose was recommended for the next cohort. The dose recommended for use in phase 2 part was analyzed on the basis of the safety and tolerability data obtained in phase 1 part of the study.
Phase 2 Part: Confirmed Objective Response Rate (ORR) as Assessed by the Independent Review Committee (IRC)
Confirmed ORR is defined as percentage of participants who were confirmed to had achieved complete response (CR) or partial response (PR) per IRC using Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 after the initiation of study treatment. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 millimeter (mm) in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in sum of the longest diameters (SLD) of target lesions, taking as reference baseline sum diameters. The SLD must also demonstrate an absolute increase of at least 5 mm.

Secondary Outcome Measures

Phase 1 Part: Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
Phase 1 Part: Number of Participants With First Cycle DLTs Based on National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 5.00
DLT was defined as drug-related toxicity which met one of the following criteria and occurred within the first 28 days of study treatment (Cycle 1): Non-hematologic toxicities any >=grade (G) 3 non-hematologic toxicity, with exception of self-limiting or medically controllable toxicities(nausea, vomiting, fatigue, electrolyte disturbances, hypersensitivity reactions) lasting <3 days, excluding alopecia. Hematologic toxicities: febrile neutropenia not related to underlying disease (fever,>38.3 degree Celsius [C]); absolute neutrophil count <0.5*10^9 per liter [/L]); prolonged G4 neutropenia (>=7 days) (if granulocyte-colony stimulating factor [G-CSF] required, event considered as DLT irrespective of the duration); neutropenic infection:>=G3 neutropenia with >=G3 infection; thrombocytopenia >=G3 with bleeding, >=G3 requiring platelet transfusion or G4 without bleeding lasting >=7 days. Missed >=25% of planned doses of drug over 28 days due to treatment-related AEs in first cycle.
Phase 1 Part: Number of DLTs for Mobocertinib Based on NCI CTCAE, Version 5.00
DLT was defined as drug-related toxicity which met one of the following criteria and occurred within the first 28 days of study treatment (Cycle 1): Non-hematologic toxicities any >=G 3 non-hematologic toxicity, with exception of self-limiting or medically controllable toxicities (nausea, vomiting, fatigue, electrolyte disturbances, hypersensitivity reactions) lasting <3 days, excluding alopecia. Hematologic toxicities: febrile neutropenia not related to underlying disease (fever,>38.3 degree C); absolute neutrophil count <0.5*10^9/L); prolonged G4 neutropenia (>=7 days) (if granulocyte-colony stimulating factor [G-CSF] required, event considered as DLT irrespective of the duration); neutropenic infection: >=G3 neutropenia with >=G3 infection; thrombocytopenia >=G3 with bleeding, >=G3 requiring platelet transfusion or G4 without bleeding lasting >=7 days. Missed >=25% of planned doses of drug over 28 days due to treatment-related AEs in first cycle.
Phase 1 Part: Maximum Tolerated Dose (MTD) of Orally Administered Mobocertinib
The MTD was declared when at least 9 participants were evaluable in the study and 6 participants were evaluable at the current dose, and the current dose was recommended for the next cohort.
Phase 1 Part, Cmax: Maximum Observed Plasma Concentration for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) After a Single Oral Dose
Phase 1 Part, Tmax: Time of First Occurrence of Cmax for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) After a Single Oral Dose
Phase 1 Part, AUC24: Area Under the Plasma Concentration-Time Curve From Time 0 to 24 Hours for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) After a Single Oral Dose
Phase 1 Part, Cmax, ss: Maximum Observed Plasma Concentration for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Phase 1 Part, Tmax, ss: Time of First Occurrence of Cmax for Mobocertinib Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Phase 1 Part, AUC24, ss: Area Under the Plasma Concentration-Time Curve From Time 0 to 24 Hours for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Phase 1 Part, Rac (AUC 24): Extent of Accumulation Ratio Based on AUC 24 on Multiple Dosing for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Rac (AUC 24) was calculated as the ratio of drug concentrations observed during a dosing interval at steady state divided by drug concentrations seen during the dosing interval after a single (first) dose. Rac (AUC 24) = AUC(0-24) on Cycle 2 Day 1/ AUC(0-24) on Cycle 1 Day 1.
Phase 1 Part: ORR in Participants With EGFR Mutations as Assessed by Investigator
Investigator assessed ORR using RECIST version 1.1 in participants with EGFR mutations. ORR was defined as the percentage of participants achieving CR and PR per RECIST version 1.1. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters. All participants with ORR had documentation of EGFR exon 20 insertion mutation.
Phase 1 Part: ORR in Participants With HER2 Mutations as Assessed by Investigator
Investigator assessed ORR using RECIST version 1.1 in participants with HER2 mutations. ORR was defined as the percentage of participants achieving CR and PR per RECIST version 1.1. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.
Phase 2 Part: Confirmed ORR as Assessed by the Investigator
Confirmed ORR was defined as the percentage of the participants who were confirmed to have achieved CR or PR per the investigator using RECIST version 1.1. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.
Phase 2 Part: Duration of Response (DOR) as Assessed by the IRC as Per RECIST V1.1
Duration of response as assessed by the IRC was defined as the time interval from first documentation of CR/PR (whichever is first recorded) until the first date that PD is objectively documented. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level; PR: At least a 30% decrease in SLD of target lesions, taking as a reference the baseline SLD. PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest); PD (non-target lesion response): Unequivocal progression of existing non-target lesions. The SLD must also demonstrate an absolute increase of at least 5 mm.
Phase 2 Part: DOR as Assessed by Investigator as Per RECIST V1.1
Duration of response as assessed by the investigator was defined as the time interval from first documentation of CR/PR (whichever is first recorded) until the first date that PD is objectively documented. CR (target lesion response):disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level; PR: At least a 30% decrease in SLD of target lesions, taking as a reference the baseline SLD. PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest); PD (non-target lesion response): Unequivocal progression of existing non-target lesions. The SLD must also demonstrate an absolute increase of at least 5 mm.
Phase 2 Part: Time to Response as Assessed by the IRC as Per RECIST V1.1
Time to response as assessed by the IRC was defined as the time interval from the date of the first dose of study treatment until the initial observation of CR or PR for participants with confirmed CR/PR. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR: at least a 30% decrease in the SLD of target lesions, taking as reference the baseline sum diameters.
Phase 2 Part: Time to Response as Assessed by the Investigator Per RECIST V1.1
Time to response as assessed by the investigator was defined as the time interval from the date of the first dose of study treatment until the initial observation of CR or PR for participants with confirmed CR/PR. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR: at least a 30% decrease in the SLD of target lesions, taking as reference the baseline sum diameters.
Phase 2 Part: Disease Control Rate (DCR) as Assessed by the IRC as Per RECIST V1.1
DCR as assessed by IRC was defined as percentage of participants achieved CR, PR, stable disease (SD) (measurements must had met SD criteria at least once after study entry at minimum interval of 42 days) after initiation of study drug.CR(target lesion): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis.CR(non-target lesion): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level.PR(target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.SD(target lesion): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD.PD(target lesion): SLD increased by at least 20% from smallest value on study, SLD must also demonstrate an absolute increase of at least 5 mm.PD (non-target lesion): unequivocal progression of existing non-target lesions.
Phase 2 Part: DCR as Assessed by the Investigator Per RECIST V1.1
DCR as assessed by investigator was defined as percentage of participants achieved CR, PR, SD (measurements must had met SD criteria at least once after study entry at minimum interval of 42 days) after initiation of study drug. CR (target lesion): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.SD (target lesion): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. PD(target lesion): SLD increased by at least 20% from smallest value on study, SLD must also demonstrate an absolute increase of at least 5 mm.PD (non-target lesion): unequivocal progression of existing non-target lesions.
Phase 2 Part: Progression Free Survival (PFS) as Assessed by the IRC as Per RECIST V1.1
PFS as assessed by the IRC was defined as the time interval from the start of study treatment until to the first documentation of PD or death due to any cause (whichever comes first) according to RECIST version 1.1.PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest). The SLD must also demonstrate an absolute increase of at least 5 mm. PD (non-target lesion response): unequivocal progression of existing non-target lesions.
Phase 2 Part: PFS as Assessed by the Investigator as Per RECIST V1.1
PFS as assessed by the investigator was defined as the time interval from the start of study treatment until to the first documentation of PD or death due to any cause (whichever comes first) according to RECIST version 1.1.PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest). The SLD must also demonstrate an absolute increase of at least 5 mm. PD (non-target lesion response): unequivocal progression of existing non-target lesions.
Phase 2 Part: Overall Survival (OS)
OS was defined as the interval from the date of the first dose of the study treatment until death due to any cause.
Phase 2 Part: Change From Baseline in Patient-reported Symptoms, Functioning, and Health-related Quality of Life (HRQoL) as Assessed by the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-C30
EORTC QLQ-C30, version 3.0 was a cancer-specific questionnaire comprised of 5 functional scales (physical, role, cognitive, emotional, and social functioning); 3 symptom scales (fatigue, pain, and nausea/vomiting); a global health status (GHS)/quality-of-life (QoL) scale; and a six single-item scales (dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial difficulties). GHS/QoL scale has 7 possible scores of responses (1=very poor to 7=excellent). All other items have 4 possible scores (1=not at all, 2=a little, 3=quite a bit, 4=very much). Raw scores converted into scale scores ranging from 0 to 100. For functional and GHS/QoL scales, higher scores represent better HRQoL (positive change from Baseline=improvement), for symptom scales lower scores represent better QoL (i.e., a low level of symptomatology/problems) (negative change from Baseline=improvement), and for six-single item scale, lower scores represent better HRQoL (negative change from Baseline=improvement).
Phase 2 Part: Change From Baseline in Patient-reported Symptoms (Particular Core Symptoms of Lung Cancer), Functioning, and HRQoL as Assessed by the EORTC Lung Cancer Module QLQ-LC13
HRQOL scores was assessed with EORTC, it is a lung cancer module QLQ-LC13, version 3.0. QLQ-LC13 included 13 questions (4-point scale where 1=Not at all [best] to 4=Very much [worst]) assessing lung cancer-associated symptoms (cough, hemoptysis, dyspnea, and site-specific pain [chest, arm or shoulder, other parts]), treatment-related side effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and use of pain medication. Subscale score range: 0 to 100. Higher symptom score = greater degree of symptom severity.

Full Information

First Posted
January 15, 2019
Last Updated
October 2, 2023
Sponsor
Takeda
search

1. Study Identification

Unique Protocol Identification Number
NCT03807778
Brief Title
A Study of Mobocertinib in Japanese Adults With Non-Small Cell Lung Cancer
Official Title
A Phase 1/2 Study of the Oral EGFR/HER2 Inhibitor TAK-788 in Japanese Non-Small Cell Lung Cancer Patients
Study Type
Interventional

2. Study Status

Record Verification Date
November 2022
Overall Recruitment Status
Active, not recruiting
Study Start Date
February 4, 2019 (Actual)
Primary Completion Date
November 8, 2021 (Actual)
Study Completion Date
March 31, 2024 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Takeda

4. Oversight

Studies a U.S. FDA-regulated Drug Product
No
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
No

5. Study Description

Brief Summary
This study is in 2 parts. Different participants will take part in the 1st and 2nd parts of the study. The main aim of the 1st part of the study is to check how much Mobocertinib adults with non-small cell lung cancer (NSCLC) can receive without getting side effects from it. The main aim of the 2nd part of the study is to learn if the condition of adults with non-small cell lung cancer improves after treatment with Mobocertinib. Another aim is to continue checking for side effects from Mobocertinib. In the 1st part of the study, at the first visit, the study doctor will check who can take part. For those that can take part, participants will take a capsule of Mobocertinib once a day for 28 days. This will count as 1 cycle. Different small groups of participants will receive lower to higher doses of Mobocertinib. The study doctors will check for side effects after each dose of TAK 788. In this way, researchers can work out the best dose of Mobocertinib to give participants in the 2nd part of the study. Participants will visit the clinic 30 days after their treatment has finished for a final check-up. In the 2nd part of the study, at the first visit, the study doctor will check who can take part. Participants will receive the best dose of Mobocertinib worked out from the 1st part of the study. Participants will receive Mobocertinib in the same way as those from the 1st part of the study. The study doctors will learn if the condition of these participants improves after treatment with Mobocertinib. The study doctors will also check for side effects from Mobocertinib. After treatment has finished, participants will visit the clinic every 12 weeks until the end of the study. In both parts of the study, participants can receive Mobocertinib for up to just over 1 year, or longer if their condition stays improved.
Detailed Description
The drug being tested in this study is called Mobocertinib. Mobocertinib is being tested to treat Japanese participants with NSCLC. This study has two parts (Phase 1 part and Phase 2 part), Phase 1 part of this study will look at the safety, efficacy, tolerability and PK of Mobocertinib orally administered once daily, and will determine a RP2D. Phase 2 study will look at the efficacy and safety of Mobocertinib in treatment naive Japanese NSCLC patients with epidermal growth factor receptor (EGFR) exon 20 insertion mutation. All participants will be assigned to Phase 1 part or Phase 2 part and will be asked to take Mobocertinib capsule as following dosage and regimen; Phase 1 part; Mobocertinib, 40 mg as starting dose, once daily, and escalating up to 160 mg until a Maximum Tolerated Dose (MTD). An expansion phase may be followed at any dose to further confirm safety observations following identification of MTD/RP2D. Phase 2 part; Mobocertinib, 160 mg, once daily The study will enroll approximately 58-63 participants (Phase 1 part; 28-33 and Phase 2 part; 30). This multi-center trial will be conducted in Japan. The overall time to participate in this study of Phase 1 part is approximately 3 years and Phase 2 part is approximately 4 years. Participants will make multiple visits to the clinic in the treatment period, and the post-treatment period including follow-up assessments after the last dose of the study drug.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Non-Small Cell Lung Cancer

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 1
Interventional Study Model
Single Group Assignment
Masking
None (Open Label)
Allocation
Non-Randomized
Enrollment
53 (Actual)

8. Arms, Groups, and Interventions

Arm Title
Mobocertinib, Phase 1 Part
Arm Type
Experimental
Arm Description
Mobocertinib 40 milligrams (mg) (as the starting dose), capsules, orally, once daily on Days 1-28 of each 28-day treatment cycle for up to disease progression or intolerable toxicity, or another discontinuation criterion, and increasing until 160 mg, once daily (for up to approximately 10-12 cycles).
Arm Title
Mobocertinib, Phase 2 Part
Arm Type
Experimental
Arm Description
Mobocertinib 160 mg, once daily, for up to approximately 10-12 cycles.
Intervention Type
Drug
Intervention Name(s)
Mobocertinib
Other Intervention Name(s)
TAK-788, AP32788
Intervention Description
Mobocertinib capsule.
Primary Outcome Measure Information:
Title
Phase 1 Part: Recommended Phase 2 Dose (RP2D) of Orally Administered Mobocertinib
Description
The RP2D was the maximum tolerated dose (MTD) or less. The MTD was declared when at least 9 participants were evaluable in the study and 6 participants were evaluable at the current dose, and the current dose was recommended for the next cohort. The dose recommended for use in phase 2 part was analyzed on the basis of the safety and tolerability data obtained in phase 1 part of the study.
Time Frame
Cycle 1 (Cycle length=28 days)
Title
Phase 2 Part: Confirmed Objective Response Rate (ORR) as Assessed by the Independent Review Committee (IRC)
Description
Confirmed ORR is defined as percentage of participants who were confirmed to had achieved complete response (CR) or partial response (PR) per IRC using Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 after the initiation of study treatment. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 millimeter (mm) in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in sum of the longest diameters (SLD) of target lesions, taking as reference baseline sum diameters. The SLD must also demonstrate an absolute increase of at least 5 mm.
Time Frame
From the first dose of the study drug until progressive disease (PD) (up to 2 years and 9 months)
Secondary Outcome Measure Information:
Title
Phase 1 Part: Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
Time Frame
From first dose of study drug until 30 days after the last dose or before initiation of new anticancer therapy (whichever comes first) (Up to 2 years and 9 months, till data cut-off of 08 November 2021)
Title
Phase 1 Part: Number of Participants With First Cycle DLTs Based on National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 5.00
Description
DLT was defined as drug-related toxicity which met one of the following criteria and occurred within the first 28 days of study treatment (Cycle 1): Non-hematologic toxicities any >=grade (G) 3 non-hematologic toxicity, with exception of self-limiting or medically controllable toxicities(nausea, vomiting, fatigue, electrolyte disturbances, hypersensitivity reactions) lasting <3 days, excluding alopecia. Hematologic toxicities: febrile neutropenia not related to underlying disease (fever,>38.3 degree Celsius [C]); absolute neutrophil count <0.5*10^9 per liter [/L]); prolonged G4 neutropenia (>=7 days) (if granulocyte-colony stimulating factor [G-CSF] required, event considered as DLT irrespective of the duration); neutropenic infection:>=G3 neutropenia with >=G3 infection; thrombocytopenia >=G3 with bleeding, >=G3 requiring platelet transfusion or G4 without bleeding lasting >=7 days. Missed >=25% of planned doses of drug over 28 days due to treatment-related AEs in first cycle.
Time Frame
Cycle 1 (Cycle length=28 days)
Title
Phase 1 Part: Number of DLTs for Mobocertinib Based on NCI CTCAE, Version 5.00
Description
DLT was defined as drug-related toxicity which met one of the following criteria and occurred within the first 28 days of study treatment (Cycle 1): Non-hematologic toxicities any >=G 3 non-hematologic toxicity, with exception of self-limiting or medically controllable toxicities (nausea, vomiting, fatigue, electrolyte disturbances, hypersensitivity reactions) lasting <3 days, excluding alopecia. Hematologic toxicities: febrile neutropenia not related to underlying disease (fever,>38.3 degree C); absolute neutrophil count <0.5*10^9/L); prolonged G4 neutropenia (>=7 days) (if granulocyte-colony stimulating factor [G-CSF] required, event considered as DLT irrespective of the duration); neutropenic infection: >=G3 neutropenia with >=G3 infection; thrombocytopenia >=G3 with bleeding, >=G3 requiring platelet transfusion or G4 without bleeding lasting >=7 days. Missed >=25% of planned doses of drug over 28 days due to treatment-related AEs in first cycle.
Time Frame
Cycle 1 (Cycle length=28 days)
Title
Phase 1 Part: Maximum Tolerated Dose (MTD) of Orally Administered Mobocertinib
Description
The MTD was declared when at least 9 participants were evaluable in the study and 6 participants were evaluable at the current dose, and the current dose was recommended for the next cohort.
Time Frame
Cycle 1 (Cycle length=28 days)
Title
Phase 1 Part, Cmax: Maximum Observed Plasma Concentration for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) After a Single Oral Dose
Time Frame
Cycle 1 Day 1: pre-dose and at 0.5 1, 2, 4, 6, 8 and 24 hours post-dose (Cycle length = 28 days)
Title
Phase 1 Part, Tmax: Time of First Occurrence of Cmax for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) After a Single Oral Dose
Time Frame
Cycle 1 Day 1: pre-dose and at 0.5 1, 2, 4, 6, 8 and 24 hours post-dose (Cycle length = 28 days)
Title
Phase 1 Part, AUC24: Area Under the Plasma Concentration-Time Curve From Time 0 to 24 Hours for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) After a Single Oral Dose
Time Frame
Cycle 1 Day 1: pre-dose and at 0.5 1, 2, 4, 6, 8 and 24 hours post-dose (Cycle length = 28 days)
Title
Phase 1 Part, Cmax, ss: Maximum Observed Plasma Concentration for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Time Frame
Cycle 2 Day 1: pre-dose and at 0.5 1, 2, 4, 6, 8 and 24 hours post-dose (Cycle length = 28 days)
Title
Phase 1 Part, Tmax, ss: Time of First Occurrence of Cmax for Mobocertinib Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Time Frame
Cycle 2 Day 1: pre-dose and at 0.5 1, 2, 4, 6, 8 and 24 hours post-dose (Cycle length = 28 days)
Title
Phase 1 Part, AUC24, ss: Area Under the Plasma Concentration-Time Curve From Time 0 to 24 Hours for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Time Frame
Cycle 2 Day 1: pre-dose and at 0.5 1, 2, 4, 6, 8 and 24 hours post-dose (Cycle length = 28 days)
Title
Phase 1 Part, Rac (AUC 24): Extent of Accumulation Ratio Based on AUC 24 on Multiple Dosing for Mobocertinib and Its Active Metabolites (AP32960 and AP32914) at Steady State After Multiple Oral Doses
Description
Rac (AUC 24) was calculated as the ratio of drug concentrations observed during a dosing interval at steady state divided by drug concentrations seen during the dosing interval after a single (first) dose. Rac (AUC 24) = AUC(0-24) on Cycle 2 Day 1/ AUC(0-24) on Cycle 1 Day 1.
Time Frame
Cycle 2 Day 1: pre-dose and at 0.5 1, 2, 4, 6, 8 and 24 hours post-dose (Cycle length = 28 days)
Title
Phase 1 Part: ORR in Participants With EGFR Mutations as Assessed by Investigator
Description
Investigator assessed ORR using RECIST version 1.1 in participants with EGFR mutations. ORR was defined as the percentage of participants achieving CR and PR per RECIST version 1.1. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters. All participants with ORR had documentation of EGFR exon 20 insertion mutation.
Time Frame
From the first dose of the study drug until PD (up to 2 years and 9 months, till data cut-off of 08 November 2021)
Title
Phase 1 Part: ORR in Participants With HER2 Mutations as Assessed by Investigator
Description
Investigator assessed ORR using RECIST version 1.1 in participants with HER2 mutations. ORR was defined as the percentage of participants achieving CR and PR per RECIST version 1.1. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.
Time Frame
From the first dose of the study drug until PD (up to 2 years and 9 months, till data cut-off of 08 November 2021)
Title
Phase 2 Part: Confirmed ORR as Assessed by the Investigator
Description
Confirmed ORR was defined as the percentage of the participants who were confirmed to have achieved CR or PR per the investigator using RECIST version 1.1. Confirmed responses were responses that persisted on repeat imaging >=4 weeks after initial response. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.
Time Frame
From the first dose of the study drug until PD (up to 2 years and 9 months, till data cut-off of 08 November 2021)
Title
Phase 2 Part: Duration of Response (DOR) as Assessed by the IRC as Per RECIST V1.1
Description
Duration of response as assessed by the IRC was defined as the time interval from first documentation of CR/PR (whichever is first recorded) until the first date that PD is objectively documented. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level; PR: At least a 30% decrease in SLD of target lesions, taking as a reference the baseline SLD. PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest); PD (non-target lesion response): Unequivocal progression of existing non-target lesions. The SLD must also demonstrate an absolute increase of at least 5 mm.
Time Frame
From first documentation of CR/PR until first PD (Up to 2 years and 9 months, till data cut-off of 08 November 2021)
Title
Phase 2 Part: DOR as Assessed by Investigator as Per RECIST V1.1
Description
Duration of response as assessed by the investigator was defined as the time interval from first documentation of CR/PR (whichever is first recorded) until the first date that PD is objectively documented. CR (target lesion response):disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level; PR: At least a 30% decrease in SLD of target lesions, taking as a reference the baseline SLD. PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest); PD (non-target lesion response): Unequivocal progression of existing non-target lesions. The SLD must also demonstrate an absolute increase of at least 5 mm.
Time Frame
From first documentation of CR/PR until first PD (Up to 2 years and 9 months, till data cut-off of 08 November 2021)
Title
Phase 2 Part: Time to Response as Assessed by the IRC as Per RECIST V1.1
Description
Time to response as assessed by the IRC was defined as the time interval from the date of the first dose of study treatment until the initial observation of CR or PR for participants with confirmed CR/PR. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR: at least a 30% decrease in the SLD of target lesions, taking as reference the baseline sum diameters.
Time Frame
From the first dose of the study drug up to first confirmed CR or PR (Up to 2 years 9 months, till data cut-off of 08 November 2021)
Title
Phase 2 Part: Time to Response as Assessed by the Investigator Per RECIST V1.1
Description
Time to response as assessed by the investigator was defined as the time interval from the date of the first dose of study treatment until the initial observation of CR or PR for participants with confirmed CR/PR. CR (target lesion response): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion response): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR: at least a 30% decrease in the SLD of target lesions, taking as reference the baseline sum diameters.
Time Frame
From the first dose of the study drug up to first confirmed CR or PR (Up to 2 years 9 months, till data cut-off of 08 November 2021)
Title
Phase 2 Part: Disease Control Rate (DCR) as Assessed by the IRC as Per RECIST V1.1
Description
DCR as assessed by IRC was defined as percentage of participants achieved CR, PR, stable disease (SD) (measurements must had met SD criteria at least once after study entry at minimum interval of 42 days) after initiation of study drug.CR(target lesion): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis.CR(non-target lesion): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level.PR(target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.SD(target lesion): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD.PD(target lesion): SLD increased by at least 20% from smallest value on study, SLD must also demonstrate an absolute increase of at least 5 mm.PD (non-target lesion): unequivocal progression of existing non-target lesions.
Time Frame
From the first dose of the study drug until PD (Up to 2 years 9 month, till data cut-off of 08 November 2021)
Title
Phase 2 Part: DCR as Assessed by the Investigator Per RECIST V1.1
Description
DCR as assessed by investigator was defined as percentage of participants achieved CR, PR, SD (measurements must had met SD criteria at least once after study entry at minimum interval of 42 days) after initiation of study drug. CR (target lesion): disappearance of all extranodal target lesions, all pathological lymph nodes must have decreased to <10 mm in short axis. CR (non-target lesion): disappearance of all extranodal nontarget lesions, all lymph nodes must be nonpathological in size (<10 mm short axis) and normalization of tumor marker level. PR (target lesions): at least 30% decrease in SLD of target lesions, taking as reference baseline sum diameters.SD (target lesion): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. PD(target lesion): SLD increased by at least 20% from smallest value on study, SLD must also demonstrate an absolute increase of at least 5 mm.PD (non-target lesion): unequivocal progression of existing non-target lesions.
Time Frame
From the first dose of the study drug until PD (Up to 2 years 9 month, till data cut-off of 08 November 2021)
Title
Phase 2 Part: Progression Free Survival (PFS) as Assessed by the IRC as Per RECIST V1.1
Description
PFS as assessed by the IRC was defined as the time interval from the start of study treatment until to the first documentation of PD or death due to any cause (whichever comes first) according to RECIST version 1.1.PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest). The SLD must also demonstrate an absolute increase of at least 5 mm. PD (non-target lesion response): unequivocal progression of existing non-target lesions.
Time Frame
From the first dose of the study drug until PD or death due to any cause (whichever comes first) (Up to 2 years and 9 months, till data cut-off of 08 November 2021
Title
Phase 2 Part: PFS as Assessed by the Investigator as Per RECIST V1.1
Description
PFS as assessed by the investigator was defined as the time interval from the start of study treatment until to the first documentation of PD or death due to any cause (whichever comes first) according to RECIST version 1.1.PD (target lesion response): SLD increased by at least 20% from the smallest value on study (including baseline, if that is the smallest). The SLD must also demonstrate an absolute increase of at least 5 mm. PD (non-target lesion response): unequivocal progression of existing non-target lesions.
Time Frame
From the first dose of the study drug until PD or death due to any cause (whichever comes first) (Up to 2 years and 9 months, till data cut-off of 08 November 2021
Title
Phase 2 Part: Overall Survival (OS)
Description
OS was defined as the interval from the date of the first dose of the study treatment until death due to any cause.
Time Frame
From the start of the study drug up to death due to any cause (Up to 2 years 9 months, till data cut-off of 08 November 2021)
Title
Phase 2 Part: Change From Baseline in Patient-reported Symptoms, Functioning, and Health-related Quality of Life (HRQoL) as Assessed by the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-C30
Description
EORTC QLQ-C30, version 3.0 was a cancer-specific questionnaire comprised of 5 functional scales (physical, role, cognitive, emotional, and social functioning); 3 symptom scales (fatigue, pain, and nausea/vomiting); a global health status (GHS)/quality-of-life (QoL) scale; and a six single-item scales (dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial difficulties). GHS/QoL scale has 7 possible scores of responses (1=very poor to 7=excellent). All other items have 4 possible scores (1=not at all, 2=a little, 3=quite a bit, 4=very much). Raw scores converted into scale scores ranging from 0 to 100. For functional and GHS/QoL scales, higher scores represent better HRQoL (positive change from Baseline=improvement), for symptom scales lower scores represent better QoL (i.e., a low level of symptomatology/problems) (negative change from Baseline=improvement), and for six-single item scale, lower scores represent better HRQoL (negative change from Baseline=improvement).
Time Frame
Baseline and at 30 days after last dose (at Month 19)
Title
Phase 2 Part: Change From Baseline in Patient-reported Symptoms (Particular Core Symptoms of Lung Cancer), Functioning, and HRQoL as Assessed by the EORTC Lung Cancer Module QLQ-LC13
Description
HRQOL scores was assessed with EORTC, it is a lung cancer module QLQ-LC13, version 3.0. QLQ-LC13 included 13 questions (4-point scale where 1=Not at all [best] to 4=Very much [worst]) assessing lung cancer-associated symptoms (cough, hemoptysis, dyspnea, and site-specific pain [chest, arm or shoulder, other parts]), treatment-related side effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and use of pain medication. Subscale score range: 0 to 100. Higher symptom score = greater degree of symptom severity.
Time Frame
Baseline and at 30 days after last dose (at Month 19)

10. Eligibility

Sex
All
Minimum Age & Unit of Time
20 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: General Inclusion Criteria (Both in Phase 1 and Phase 2 Part); Male or female patients ≥20 years old. Must have measurable disease by RECIST v1.1. Previously irradiated lesions may not be used for target lesions, unless there is unambiguous radiological progression after radiotherapy. Eastern Cooperative Oncology Group (ECOG) performance status 0 to 1. Minimum life expectancy of 3 months or more. Adequate renal and hepatic function as defined by the following criteria: •Total serum bilirubin ≤1.5 × upper limit of normal (ULN) (≤3.0 × ULN for patients with Gilbert syndrome or if liver function abnormalities are due to underlying malignancy); •Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) ≤2.5 × ULN (or ≤5 × ULN if liver function abnormalities are due to underlying malignancy); •Estimated creatinine clearance ≥30 mL/min (calculated by using the Cockcroft-Gault equation); •Serum albumin ≥2 g/dL; and •Serum lipase ≤1.5 × ULN; and •Serum amylase ≤1.5 × ULN unless the increased serum amylase is due to salivary isoenzymes. Adequate bone marrow function as defined by the following criteria: Absolute neutrophil count ≥1.5 × 109/L; Platelet count ≥75 × 109/L in Phase 1 Part and ≥100 × 109/L in Phase 2 Part; and Hemoglobin ≥9.0 g/dL. Normal QT interval on screening ECG, defined as QTcF of ≤450 ms in males or ≤470 ms in females. Female patients who: Are postmenopausal (natural amenorrhea and not due to other medical reasons) for at least 1 year before the screening visit, OR Are surgically sterile, OR If they are of childbearing potential, agree to practice 1 highly effective non-hormonal method of contraception and 1 additional effective (barrier) method (see Section 8.7.1) at the same time, from the time of signing the informed consent form (ICF) through 30 days after the last dose of study drug, OR Agree to practice true abstinence, when this is in line with the preferred and usual lifestyle of the subject. Note: Periodic abstinence (eg, calendar, ovulation, symptothermal, postovulation methods), withdrawal, spermicides only, and lactational amenorrhea are not acceptable methods of contraception. Male patients, even if surgically sterilized (ie, status postvasectomy), who: Agree to practice effective barrier contraception during the entire study treatment period and through 30 days after the last dose of study drug, OR Agree to practice true abstinence, when this is in line with the preferred and usual lifestyle of the subject. Note: Periodic abstinence (eg, calendar, ovulation, symptothermal, postovulation methods), withdrawal, spermicides only, and lactational amenorrhea are not acceptable methods of contraception. 9.Voluntary written consent must be given before performance of any study related procedure not part of standard medical care, with the understanding that consent may be withdrawn by the patient at any time without prejudice to future medical care. 10.Willingness and ability to comply with scheduled visits and study procedures. Phase-Specific Inclusion Criteria (Phase 1 part); 1.Have histologically or cytologically confirmed locally advanced (and not a candidate for definitive therapy) (Stage IIIB) or metastatic NSCLC (Stage IV). 2.Refractory to standard available therapies. 3.All toxicities from prior therapy have resolved to ≤ grade 1 according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE v5.0), or have resolved to baseline, at the time of first dose of Mobocertinib. Note: treatment-related grade >1 alopecia or treatment-related grade 2 peripheral neuropathy are allowed if deemed irreversible. Phase-Specific Inclusion Criteria (Phase 2 part); Histologically or cytologically confirmed locally advanced not suitable for definitive therapy, recurrent, or metastatic (Stage IV) NSCLC. Not received prior systemic treatment for locally advanced or metastatic disease (with the exception below): Neoadjuvant or adjuvant chemotherapy/immunotherapy for Stage I to III or combined modality chemotherapy/radiation for locally advanced disease is allowed if completed >6 months before the development of metastatic disease. A documented EGFR in-frame exon 20 insertion (including A763_Y764insFQEA, V769_D770insASV, D770_N771insNPG, D770_N771insSVD, H773_V774insNPH, or any other in-frame exon 20 insertion mutation) by a local test that has been analytically validated per local authority guidelines. The EGFR exon 20 insertion mutation can be either alone or in combination with other EGFR or HER2 mutations except EGFR common mutations (exon 19 del or L858R). Adequate tumor tissue available, either from primary or metastatic sites, for central laboratory confirmation of EGFR in-frame exon 20 insertion mutation. Note: confirmation of central test positivity is not required before the first dose of Mobocertinib. Exclusion Criteria: General Exclusion Criteria (Both in Phase 1 and Phase 2 Part); Have been diagnosed with another primary malignancy other than NSCLC except for adequately treated non-melanoma skin cancer or cervical cancer in situ; definitively treated non-metastatic prostate cancer; or patients with another primary malignancy who are definitively relapse-free with at least 3 years elapsed since the diagnosis of the other primary malignancy. Have undergone major surgery within 28 days prior to first dose of Mobocertinib. Minor surgical procedures, such as catheter placement or minimally invasive biopsy, are allowed. Have current spinal cord compression (symptomatic or asymptomatic and detected by radiographic imaging) or leptomeningeal disease (symptomatic or asymptomatic). Have significant, uncontrolled, or active cardiovascular disease, including, but not restricted to: Myocardial infarction within 6 months prior to the first dose of study drug; Unstable angina within 6 months prior to first dose; Congestive heart failure within 6 months prior to first dose; History of clinically significant (as determined by the treating physician) atrial arrhythmia; Any history of ventricular arrhythmia; or Cerebrovascular accident or transient ischemic attack within 6 months prior to first dose. 5.Have uncontrolled hypertension. Patients with hypertension should be under treatment on study entry to control blood pressure. 6.Currently being treated with medications known to be associated with the development of Torsades de Pointes. 7.Have an ongoing or active infection, including, but not limited to, the requirement for intravenous antibiotics. Have a known history of HIV infection. Testing of HIV is not required in the absence of history.Hepatitis B surface antigen (HBsAg) positive patients are allowed to enroll if hepatitis B virus (HBV)-DNA is below 1000 copies/mL in the plasma.Patients who have positive hepatitis C virus (HCV) antibody can be enrolled but must have HCV-RNA undetectable in the plasma. 8.Currently have or have a history of interstitial lung disease (ILD), radiation pneumonitis that required steroid treatment, or drug-related pneumonitis. 9.Female patients who are lactating and breastfeeding or have a positive serum pregnancy test during the screening period. Note: Female patients who are lactating will be eligible if they discontinue breastfeeding. 10.Have gastrointestinal illness or disorder that could affect oral absorption of Mobocertinib. 11.Have any condition or illness that, in the opinion of the investigator, might compromise patient safety or interfere with the evaluation of the safety of the drug. Phase-Specific Exclusion Criteria (Phase 1 part); Previously received Mobocertinib. Received small-molecule anticancer therapy (including cytotoxic chemotherapy and investigational agents) within 14 days prior to the first dose of Mobocertinib (except for reversible EGFR TKIs [ie, erlotinib or gefitinib] up to 7 days prior to the first dose of Mobocertinib). Received antineoplastic monoclonal antibodies including immunotherapy within 28 days prior to the first dose of Mobocertinib. Received radiotherapy within 14 days prior to the first dose of Mobocertinib, Stereotactic radiosurgery (SRS) and stereotactic body radiosurgery are allowed up to 7 days prior to the first dose. Have symptomatic CNS metastases (parenchymal or leptomeningeal) at screening or asymptomatic disease requiring corticosteroids to control symptoms within 7 days prior to the first dose of Mobocertinib. Note: If a patient has worsening neurological symptoms or signs due to CNS metastases, the patient needs to complete local therapy and be neurologically stable (with no requirement for corticosteroids or use of anticonvulsants) for 7 days prior to the first dose of Mobocertinib. Patients with no prior history of signs or symptoms of CNS metastases but who receive prophylactic steroids or anticonvulsants are allowed. Received a strong cytochrome P450 (CYP)3A inhibitor or strong CYP3A inducer within 2 weeks prior to first dose of Mobocertinib. Phase-Specific Exclusion Criteria (Phase 2 part); Received radiotherapy within 14 days before the first dose of Mobocertinib or has not recovered from radiotherapy-related toxicities. Stereotactic radiosurgery, stereotactic body radiotherapy, or palliative radiation outside the chest and brain is allowed up to 7 days before the first dose of Mobocertinib. Have known active brain metastases (have either previously untreated intracranial CNS metastases or previously treated intracranial CNS metastases with radiologically documented new or progressing CNS lesions). Brain metastases are allowed if they have been treated with surgery and/or radiation and have been stable without requiring corticosteroids to control symptoms within 7 days before the first dose of Mobocertinib, and have no evidence of new or enlarging brain metastases. Received a moderate or strong CYP3A inhibitor or moderate or strong CYP3A inducer within 10 days prior to first dose of Mobocertinib. Have cardiac ejection fraction <50% by echocardiogram or multigated acquisition (MUGA) scan at screening.
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Study Director
Organizational Affiliation
Takeda
Official's Role
Study Director
Facility Information:
Facility Name
Aichi Cancer Center Hospital
City
Nagoya
State/Province
Aichi
Country
Japan
Facility Name
Fujita Health University Hospital
City
Toyoake
State/Province
Aichi
Country
Japan
Facility Name
National Cancer Center Hospital East
City
Kashiwa
State/Province
Chiba
Country
Japan
Facility Name
Kurume University Hospital
City
Kurume
State/Province
Fukuoka
Country
Japan
Facility Name
Hokkaido Cancer Center
City
Sapporo
State/Province
Hokkaido
Country
Japan
Facility Name
Hyogo Cancer Cente
City
Akashi
State/Province
Hyogo
Country
Japan
Facility Name
Kanazawa University Hospital
City
Kanazawa
State/Province
Ishikawa
Country
Japan
Facility Name
Kanagawa Cancer Center
City
Yokohama
State/Province
Kanagawa
Country
Japan
Facility Name
Sendai Kousei Hospital
City
Sendai
State/Province
Miyagi
Country
Japan
Facility Name
Okayama University Hospital
City
Kita-ku
State/Province
Okayama
Country
Japan
Facility Name
Kansai Medical University Hospital
City
Hirakata
State/Province
Osaka
Country
Japan
Facility Name
Kindai University Hospital
City
Osakasayama
State/Province
Osaka
Country
Japan
Facility Name
National Hospital Organization Kinki-Chuo Chest Medical Center
City
Sakai
State/Province
Osaka
Country
Japan
Facility Name
Saitama Cancer Center
City
Ina
State/Province
Saitama
Country
Japan
Facility Name
Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital
City
Bunkyo-ku
State/Province
Tokyo
Country
Japan
Facility Name
National Cancer Center Hospital
City
Chuo-ku
State/Province
Tokyo
Country
Japan
Facility Name
Cancer Institute Hospital
City
Koto-ku
State/Province
Tokyo
Country
Japan
Facility Name
Yamaguchi Ube Medical Center
City
Ube
State/Province
Yamaguchi
Country
Japan
Facility Name
Kyushu University Hospital
City
Fukuoka
Country
Japan
Facility Name
Hiroshima University Hospital
City
Hiroshima
Country
Japan
Facility Name
Kyoto University Hospital
City
Kyoto
Country
Japan
Facility Name
Niigata Cancer Center Hospital
City
Niigata
Country
Japan
Facility Name
Osaka International Cancer Institute
City
Osaka
Country
Japan
Facility Name
Tokushima University Hospital
City
Tokushima
Country
Japan
Facility Name
Wakayama Medical University Hospital
City
Wakayama
Country
Japan

12. IPD Sharing Statement

Plan to Share IPD
Yes
IPD Sharing Plan Description
Takeda provides access to the de-identified individual participant data (IPD) for eligible studies to aid qualified researchers in addressing legitimate scientific objectives (Takeda's data sharing commitment is available on https://clinicaltrials.takeda.com/takedas-commitment?commitment=5). These IPDs will be provided in a secure research environment following approval of a data sharing request, and under the terms of a data sharing agreement.
IPD Sharing Access Criteria
IPD from eligible studies will be shared with qualified researchers according to the criteria and process described on https://vivli.org/ourmember/takeda/. For approved requests, the researchers will be provided access to anonymized data (to respect patient privacy in line with applicable laws and regulations) and with information necessary to address the research objectives under the terms of a data sharing agreement.
IPD Sharing URL
https://vivli.org/ourmember/takeda/
Links:
URL
https://clinicaltrials.takeda.com/study-detail/5f6b603a4db2bf003ab4a2bd
Description
To obtain more information on the study, click here/on this link

Learn more about this trial

A Study of Mobocertinib in Japanese Adults With Non-Small Cell Lung Cancer

We'll reach out to this number within 24 hrs