Efficacy, Safety, and Pharmacokinetic of MSC2156119J in Asian Participants With Hepatocellular Carcinoma

Sponsor
Merck KGaA, Darmstadt, Germany (Industry)
Overall Status
Completed
CT.gov ID
NCT01988493
Collaborator
(none)
117
43
5
82.9
2.7
0

Study Details

Study Description

Brief Summary

This is an open-label, integrated, Phase 1b/2 trial to determine the recommended Phase 2 dose (RP2D) and to evaluate the efficacy, safety, and pharmacokinetic of MSC2156119J as first-line treatment versus sorafenib in subjects with MET+, Barcelona Clinic Liver Cancer (BCLC) Stage C, systemic treatment naive advanced hepatocellular carcinoma (HCC) and Child-Pugh class A liver function.

Condition or Disease Intervention/Treatment Phase
Phase 1/Phase 2

Study Design

Study Type:
Interventional
Actual Enrollment :
117 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
A Multicenter, Randomized, Phase Ib/II Trial to Evaluate the Efficacy, Safety, and Pharmacokinetics of MSC2156119J as Monotherapy Versus Sorafenib in Asian Subjects With MET+ Advanced Hepatocellular Carcinoma and Child-Pugh Class A Liver Function
Actual Study Start Date :
Jan 6, 2014
Actual Primary Completion Date :
Feb 5, 2018
Actual Study Completion Date :
Dec 3, 2020

Arms and Interventions

Arm Intervention/Treatment
Experimental: Phase 1b: Tepotinib 300 mg

Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Drug: Tepotinib 300 mg
Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Experimental: Phase 1b: Tepotinib 500 mg

Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Drug: Tepotinib 500 mg
Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Experimental: Phase 1b: Tepotinib 1000 mg

Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Drug: Tepotinib 1000 mg
Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal

Experimental: Phase 2: Tepotinib

Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Drug: Tepotinib
Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Experimental: Phase 2 Sorafenib

Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Drug: Sorafenib
Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.

Outcome Measures

Primary Outcome Measures

  1. Phase 1b: Number of Participants Experiencing Dose Limiting Toxicity [Day 1 to Day 21 of Cycle 1 (each cycle is 21 days)]

    Dose limiting toxicity (DLT) was defined as toxicities at any dose level and judged to be related to the study treatment by investigator and/or the sponsor. DLTs included Grade 4 neutropenia for more than 7 days; Grade greater than or equal to (>=) 3 febrile neutropenia for more than 1 day; Grade 4 thrombocytopenia or Grade 3 thrombocytopenia with non-traumatic bleeding; Grade >= 3 uncontrolled nausea/vomiting and/or diarrhea despite adequate and optimal treatment and Grade >= 3 any non-hematological adverse event (AE), except the aforementioned gastrointestinal events and alopecia. Number of participants who experienced DLT during phase 1b were reported.

  2. Phase 1b: Number of Participants With Treatment Emergent Adverse Events (TEAEs) and Serious TEAEs [Baseline up to Day 30 after the last dose of study treatment, assessed up to 94 weeks]

    An AE was defined as any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. A serious AE was an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. Treatment-emergent are events between first dose of study drug and assessed up to 94 weeks. TEAEs include both Serious TEAEs and non-serious TEAEs.

  3. Phase 2: Time to Progression (TTP) Based on Tumor Assessment by an Independent Review Committee (IRC) [From randomization to date of the observation of radiological progressive disease, assessed up to maximum 3.8 years]

    TTP was defined as the time in months from randomization to date of the observation of radiological progressive disease (PD) (based on Response Evaluation Criteria in Solid Tumors [RECIST] v1.1) assessed by an IRC. PD is defined as at least 20 percent (%) increase in sum of diameters of target lesions, taking as reference as smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must demonstrate an absolute increase of at least 5 millimeter (mm).

Secondary Outcome Measures

  1. Phase 2: Progression Free Survival (PFS) Time Based on Tumor Assessment by the Independent Review Committee (IRC) [Up to 2.8 years]

    Progression-free survival (PFS) time was defined as the time in months from randomization to either first observation of disease progression (based on RECIST v1.1) or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 millimeter(mm). PFS was measured using Kaplan-Meier (KM) estimates.

  2. Phase 2: Overall Survival (OS) [Time from randomization to the date of death or up to 6.9 years]

    Overall survival time was measured as time in months between the date of randomization and the date of death.

  3. Phase 2: Time to Progression (TTP) Based on Tumor Assessment by Investigator [From randomization to date of the observation of radiological progressive disease, assessed up to maximum 2.8 years]

    TTP was defined as the time in months from randomization to date of the observation of radiological PD (based on RECIST v1.1) assessed by the investigator. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm.

  4. Phase 1b: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-infinity) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    The AUC(0-inf) was estimated by determining the total area under the curve of the concentration versus time curve extrapolated to infinity.

  5. Phase 1b: Area Under the Plasma Concentration-Time Curve From Time Zero to the Last Sampling Time (AUC 0-t) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    Area under the plasma concentration versus time curve from time zero to the last sampling time t at which the concentration is at or above the lower limit of quantification (LLLQ). AUC(0-t) was calculated according to the mixed log-linear trapezoidal rule.

  6. Phase 1b: Area Under the Plasma Concentration-Time Curve Within 1 Dosing Interval (AUC 0-tau) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    AUC (0-tau) is the area under the plasma concentration time curve within 1 dosing interval.

  7. Phase 1b: Maximum Observed Plasma Concentration (Cmax) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    Cmax is the maximum observed plasma concentration obtained directly from the concentration versus time curve.

  8. Phase 1b: Minimum Observed Plasma Concentration (Cmin) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 15 of Cycle 1 (each Cycle is 21 days)]

    Cmin is minimum observed plasma concentration obtained directly from the concentration versus time curve.

  9. Phase 1b: Average Observed Plasma Concentration (Cav) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 15 of Cycle 1 (each Cycle is 21 days)]

    Cavg is the average plasma concentration within 1 dosing interval obtained directly from the concentration versus time curve.

  10. Phase 1b: Time to Reach Maximum Plasma Concentration (Tmax) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    Tmax is time to reach maximum observed plasma concentration obtained directly from the concentration versus time curve.

  11. Phase 1b: Apparent Volume of Distribution During the Terminal Phase (Vz/f) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    The Vz/f was defined as the theoretical volume in which the total amount of required to uniformly distribute to produce the desired plasma concentration. Apparent volume of distribution after oral dose (Vz/F) was influenced by the fraction absorbed. The Vz/f was calculated by dividing the dose with area under the concentration time curve from time zero to infinity multiplied with terminal elimination rate constant Lambda(z). Vz/f=Dose/AUC(0-inf)* Lambda(z).

  12. Phase 1b: Apparent Total Body Clearance From Plasma (CL/f) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    The CL/f is a measure of the rate at which it was metabolized or eliminated by normal biological processes. Clearance obtained after oral dose was influenced by the fraction of the dose absorbed. The CL/F from plasma was calculated using the formula: Dose divided by area under the concentration time curve from time zero to infinity (AUC0-inf).

  13. Phase 1b: Apparent Volume of Distribution During the Steady State (Vss/f) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    Volume of distribution was defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired plasma concentration of a drug. Vss/f after oral dose was influenced by the fraction absorbed.

  14. Phase 1b: Apparent Terminal Elimination Rate Constant Lambda(z) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    Lambda(z) was determined from the terminal slope of the log-transformed plasma concentration curve using linear regression method.

  15. Phase 1b: Apparent Terminal Half-life (t1/2) of Tepotinib [Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)]

    Apparent terminal half-life was defined as the time required for the plasma concentration of drug to decrease 50 percent in the final stage of its elimination.

  16. Phase 2: Time-to-Symptomatic Progression (TTSP) [Up to 6.9 years]

    Time-to-symptomatic progression was defined as time (in months) from first study drug administration to the date of deterioration of symptoms assessed by Functional Assessment of Cancer Therapy Hepatobiliary Symptom Index 8 (FHSI-8) (defined as at least a 4-point increase, i.e., higher score, compared with baseline value), or deterioration to Eastern Cooperative Oncology Group (ECOG) performance score 4, or death. FHSI-8 assesses hepatobiliary cancer symptoms with total score ranges from 0 to 32 (0 = the best quality of life; 32 = the worst quality of life with severe symptoms). ECOG assess participant's performance status on a scale of 0 to 5, where 0=fully active and 5=dead.

  17. Phase 2: Objective Response Rate (ORR) Based on Tumor Assessment by the IRC [Time from randomization until the first occurrence of PD assessed up to 6.9 years]

    The objective response rate (ORR) was defined as the percentage of participants who had achieved complete response (CR) or partial response (PR) as the best overall response according to radiological assessments as adjudicated by the IRC from randomization until the first occurrence of PD. CR: Complete Response (CR) defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Partial response (PR) defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started.

  18. Phase 2: Percentage of Participants With Disease Control Based on Tumor Assessment by IRC According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria [Time from randomization until the first occurrence of PD assessed up to 6.9 years]

    Disease control was defined as CR, PR, or stable disease (SD) as the best overall response according to radiological assessments as adjudicated by the IRC from randomization until the first occurrence of PD. CR defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD was defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started. Percentage of participants with disease control were reported.

  19. Phase 2: Progression-free Survival (PFS) Based on Tumor Assessment by the Investigator [Time from randomization to disease progression or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment, assessed up to 6.9 years]

    Progression-free survival (assessed by the Investigator) time was defined as the time in months from randomization to either first observation of radiologically confirmed progression disease by the investigator or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. PFS was measured using Kaplan-Meier (KM) estimates.

  20. Phase 2: Objective Response Rate (ORR) Based on Tumor Assessment by the Investigator [Time from randomization until the first occurrence of PD assessed up to 6.9 years]

    The objective response rate was defined as the percentage of participants who had achieved CR or PR as the best overall response according to radiological assessments as adjudicated by the investigator from randomization until the first occurrence of PD. CR defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD was defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started.

  21. Phase 2: Percentage of Participants With Disease Control Based on Tumor Assessment by Investigator According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria [Approximately up to 6.9 years]

    Disease control was defined as CR, PR, or stable disease (SD) as the best overall response according to radiological assessments as adjudicated by the IRC from randomization until the first occurrence of PD. CR defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD was defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started. Percentage of participants with disease control were reported.

Eligibility Criteria

Criteria

Ages Eligible for Study:
18 Years and Older
Sexes Eligible for Study:
All
Accepts Healthy Volunteers:
No
Inclusion Criteria:
  • Histologically or cytologically confirmed HCC

  • Participants were either intermediate HCC of BCLC Stage B, who were not eligible for surgical and/or local-regional therapies or who had progressive disease (PD) after surgical and/or local-regional therapies (note: the local-regional therapy must not contain sorafenib), or advanced HCC of BCLC Stage C

  • Participants who had disease progression on or were intolerant to the prior standard treatment for advanced HCC (phase Ib Korean subjects only)

  • A tumor biopsy was required for determining MET status

  • MET+ status (Phase 2 only), as determined by the central laboratory (Phase 1b retrospectively, Phase 2 for participant selection) were defined in the protocol

  • Child-Pugh class A with no encephalopathy according to the screening assessment

  • Asian male or female, 18 years of age or older

  • Measurable disease in accordance with RECIST v1.1 (Phase 2 only)

  • Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 0 to 2

  • Eligible for treatment with sorafenib, was assessed by investigators according to the Package Insert and clinical judgment (Phase 2 only)

  • Signed and dated informed consent indicating that the participants had been informed of all the pertinent aspects of the trial prior to enrollment

  • Willingness and ability to comply with scheduled visits, treatment plans, laboratory tests, and other trial procedures

  • Life expectancy was judged by the investigator of at least 3 months

Exclusion Criteria:
  • Prior systemic anticancer treatment for advanced HCC, included targeted therapy (for example, sorafenib), chemotherapy, or any other investigational agent (Phase 2 only)

  • Prior treatment with any agent targeting the hepatocyte growth factor (HGF)/c-Met pathway

  • Prior local-regional therapy within 4 weeks prior to Day 1 of trial treatment

  • Prior history of liver transplant

  • Laboratory index at baseline were defined in the protocol

  • Past or current history of neoplasm other than HCC, except for curatively treated non-melanoma skin cancer, in situ carcinoma of the cervix, or other cancer curatively treated and with no evidence of disease for at least 5 years

  • Known central nervous system (CNS) or brain metastasis that is either symptomatic or untreated

  • Medical history of difficulty swallowing, malabsorption, or other chronic gastrointestinal disease, or conditions that may hamper compliance and/or absorption of the tested products

  • Clinically significant gastrointestinal bleeding within 4 weeks before trial entry

  • Peripheral neuropathy Grade greater than or equal to 2 (Common Terminology Criteria for Adverse Events [CTCAE] v4.0)

  • Impaired cardiac function was defined in the protocol

  • Hypertension uncontrolled by standard therapies

  • Participants with a family history of long QT syndrome or who take any agent that is known to prolong QT/QTc interval

  • Known human immunodeficiency virus (HIV) infection

  • Particpants who had acute pancreatitis and/or chronic pancreatitis, with elevated lipase and/or amylase, clinical symptoms, and/or imaging studies that are indicative of the diagnosis (Mainland Chinese participants only)

  • Known or suspected drug hypersensitivity to any ingredients of sorafenib (Phase 2 only) and MSC2156119J

  • Female participants who were pregnant or lactating, or males and females of reproductive potential not willing or not able to employ a highly effective method of birth control/contraception to prevent pregnancy from 2 weeks before receiving study drug until 3 months after receiving the last dose of study drug

  • Concurrent treatment with a non-permitted drug

  • Substance abuse, other acute or chronic medical or psychiatric condition, or laboratory abnormalities that may increase the risk associated with trial participation in the opinion of the investigator

  • Participation in another clinical trial within the past 28 days

  • Previous anticancer treatment-related toxicities not recovered to baseline or Grade 0-1 (except alopecia and peripheral neuropathy)

  • Participants with any concurrent medical condition or disease that will potentially compromise the conduct of the study at the discretion of the investigators

Contacts and Locations

Locations

Site City State Country Postal Code
1 307 Hosptial of PLA Beijing Beijing China 100071
2 Peking University Cancer Hospital Beijing Beijing China 100142
3 Fuzhou General Hospital of Nanjing Military Area Command of Chinese PLA Fuzhou Fujian China 350025
4 Sun Yat-sen University, Cancer Center Guangzhou Guangdong China 510060
5 Nanfang Hospital Guangzhou Guangdong China 510515
6 Harbin Medical University Cancer Hospital Harbin Heilongjiang China 150081
7 The Third Xiangya Hospital of Central South University Changsha Hunan China 410013
8 Nanjing Bayi Hospital Nanjing Jiangsu China 210002
9 Nanjing First Hospital Affiliated to Nanjing Medical University Nanjing Jiangsu China 210029
10 Jilin Cancer Hospital Changchun Jilin China 130012
11 Shanghai Cancer Hospital, Fudan University Shanghai Shanghai China 200032
12 Zhongshan Hospital Fudan University Shanghai Shanghai China 200032
13 Cancer Hospital, Tianjin Medical University Tianjin Zhejiang China 300060
14 Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine Zhejiang Zhejiang China 310016
15 Beijing Friendship Hospital, Capital Medical University Beijing China 100050
16 Dong-A University Hospital Busan Korea, Republic of 602-715
17 Pusan National University Hospital Busan Korea, Republic of 602-739
18 Keimyung University Dongsan Hospital Daegu Korea, Republic of 41931
19 Kyungpook National University Hospital Daegu Korea, Republic of 700-721
20 National Cancer Center Goyang-si Korea, Republic of 10408
21 CHA Bundang Medical Center, CHA University Seongnam-si, Korea, Republic of 13496
22 Seoul National University Bundang Hospital Seongnam-si Korea, Republic of 13620
23 Kyung Hee University Hospital Seoul Korea, Republic of 02447
24 Korea University Anam Hospital Seoul Korea, Republic of 02841
25 Severance Hospital, Yonsei University Seoul Korea, Republic of 03722
26 Asan Medical Center Seoul Korea, Republic of 05505
27 Gangnam Severance Hospital, Yonsei University Health System Seoul Korea, Republic of 06273
28 Samsung Medical Center Seoul Korea, Republic of 06351
29 The Catholic University of Korea, Seoul St. Mary's Hospital Seoul Korea, Republic of 06591
30 Korea University Guro Hospital Seoul Korea, Republic of 08308
31 Seoul National University Hospital Seoul Korea, Republic of 110744
32 Ajou University Hospital Suwon-si Korea, Republic of 443-380
33 Pusan National University Yangsan Hospital Yangsan Korea, Republic of 626-770
34 Changhua Christian Hospital Changhua Taiwan 50004
35 Kaohsiung Medical University Chung-Ho Memorial Hospital Kaohsiung Taiwan 807
36 Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan 83301
37 Taichung Veterans General Hospital Taichung Taiwan 40705
38 National Cheng Kung University Hospital Tainan Taiwan 704
39 Chi Mei Medical Center, Liou Ying Tainan Taiwan 736
40 National Taiwan University Hospital Taipei Taiwan 100
41 Mackay Memorial Hospital Taipei Taiwan 10449
42 Taipei Veterans General Hospital Taipei Taiwan 11217
43 Chang Gung Memorial Hospital, Linkou Taoyuan Taiwan 333

Sponsors and Collaborators

  • Merck KGaA, Darmstadt, Germany

Investigators

  • Study Director: Medical Responsible, Merck KGaA, Darmstadt, Germany

Study Documents (Full-Text)

More Information

Additional Information:

Publications

None provided.
Responsible Party:
Merck KGaA, Darmstadt, Germany
ClinicalTrials.gov Identifier:
NCT01988493
Other Study ID Numbers:
  • 200095-004
First Posted:
Nov 20, 2013
Last Update Posted:
Mar 7, 2022
Last Verified:
Dec 1, 2021
Studies a U.S. FDA-regulated Drug Product:
No
Studies a U.S. FDA-regulated Device Product:
No
Keywords provided by Merck KGaA, Darmstadt, Germany
Additional relevant MeSH terms:

Study Results

Participant Flow

Recruitment Details First participant enrolled 06 Jan 2014. Last participant last visit: 03 Dec 2020.
Pre-assignment Detail A total of 27 participants were enrolled in Phase 1b part of the study and a total of 90 participants were enrolled in phase 2 part of the study. Participants enrolled in phase 1b were not eligible for randomization in phase 2.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Period Title: Overall Study
STARTED 7 14 6 45 45
Treated 7 14 6 45 44
Safety Population 7 14 6 45 44
COMPLETED 7 14 6 45 44
NOT COMPLETED 0 0 0 0 1

Baseline Characteristics

Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg Phase 2: Tepotinib Phase 2 Sorafenib Total
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Total of all reporting groups
Overall Participants 7 14 6 45 44 116
Age (Count of Participants)
<=18 years
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
Between 18 and 65 years
6
85.7%
11
78.6%
5
83.3%
34
75.6%
37
84.1%
93
80.2%
>=65 years
1
14.3%
3
21.4%
1
16.7%
11
24.4%
7
15.9%
23
19.8%
Sex: Female, Male (Count of Participants)
Female
1
14.3%
2
14.3%
1
16.7%
4
8.9%
3
6.8%
11
9.5%
Male
6
85.7%
12
85.7%
5
83.3%
41
91.1%
41
93.2%
105
90.5%
Ethnicity (NIH/OMB) (Count of Participants)
Hispanic or Latino
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
Not Hispanic or Latino
7
100%
14
100%
6
100%
45
100%
44
100%
116
100%
Unknown or Not Reported
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
Race (NIH/OMB) (Count of Participants)
American Indian or Alaska Native
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
Asian
7
100%
14
100%
6
100%
45
100%
44
100%
116
100%
Native Hawaiian or Other Pacific Islander
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
Black or African American
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
White
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
More than one race
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%
Unknown or Not Reported
0
0%
0
0%
0
0%
0
0%
0
0%
0
0%

Outcome Measures

1. Primary Outcome
Title Phase 1b: Number of Participants Experiencing Dose Limiting Toxicity
Description Dose limiting toxicity (DLT) was defined as toxicities at any dose level and judged to be related to the study treatment by investigator and/or the sponsor. DLTs included Grade 4 neutropenia for more than 7 days; Grade greater than or equal to (>=) 3 febrile neutropenia for more than 1 day; Grade 4 thrombocytopenia or Grade 3 thrombocytopenia with non-traumatic bleeding; Grade >= 3 uncontrolled nausea/vomiting and/or diarrhea despite adequate and optimal treatment and Grade >= 3 any non-hematological adverse event (AE), except the aforementioned gastrointestinal events and alopecia. Number of participants who experienced DLT during phase 1b were reported.
Time Frame Day 1 to Day 21 of Cycle 1 (each cycle is 21 days)

Outcome Measure Data

Analysis Population Description
Dose Limiting Toxicity (DLT) set included all participants who experienced a DLT during Cycle 1, or received at least 80 percent of all planned doses of treatment during Cycle.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 6 12 5
Count of Participants [Participants]
0
0%
0
0%
0
0%
2. Primary Outcome
Title Phase 1b: Number of Participants With Treatment Emergent Adverse Events (TEAEs) and Serious TEAEs
Description An AE was defined as any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. A serious AE was an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. Treatment-emergent are events between first dose of study drug and assessed up to 94 weeks. TEAEs include both Serious TEAEs and non-serious TEAEs.
Time Frame Baseline up to Day 30 after the last dose of study treatment, assessed up to 94 weeks

Outcome Measure Data

Analysis Population Description
The safety analysis set included all participants who had received any dose of the study medication.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Any TEAE
7
100%
14
100%
6
100%
Any Serious TEAE
2
28.6%
9
64.3%
4
66.7%
3. Primary Outcome
Title Phase 2: Time to Progression (TTP) Based on Tumor Assessment by an Independent Review Committee (IRC)
Description TTP was defined as the time in months from randomization to date of the observation of radiological progressive disease (PD) (based on Response Evaluation Criteria in Solid Tumors [RECIST] v1.1) assessed by an IRC. PD is defined as at least 20 percent (%) increase in sum of diameters of target lesions, taking as reference as smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must demonstrate an absolute increase of at least 5 millimeter (mm).
Time Frame From randomization to date of the observation of radiological progressive disease, assessed up to maximum 3.8 years

Outcome Measure Data

Analysis Population Description
The modified intent-to treat (mITT) analysis set in the Phase 2 part of this study included all participants with Mesenchymal-epithelial transition (MET)+ hepatocelluar carcinoma (HCC) who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Median (90% Confidence Interval) [Months]
2.9
1.4
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.0087
Comments
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.46
Confidence Interval (2-Sided) 90%
0.28 to 0.76
Parameter Dispersion Type:
Value:
Estimation Comments
4. Secondary Outcome
Title Phase 2: Progression Free Survival (PFS) Time Based on Tumor Assessment by the Independent Review Committee (IRC)
Description Progression-free survival (PFS) time was defined as the time in months from randomization to either first observation of disease progression (based on RECIST v1.1) or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 millimeter(mm). PFS was measured using Kaplan-Meier (KM) estimates.
Time Frame Up to 2.8 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Median (90% Confidence Interval) [Months]
2.8
1.4
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.0229
Comments
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.53
Confidence Interval () 90%
0.33 to 0.84
Parameter Dispersion Type:
Value:
Estimation Comments
5. Secondary Outcome
Title Phase 2: Overall Survival (OS)
Description Overall survival time was measured as time in months between the date of randomization and the date of death.
Time Frame Time from randomization to the date of death or up to 6.9 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Median (90% Confidence Interval) [Months]
9.3
8.6
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.2333
Comments
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.71
Confidence Interval () 90%
0.45 to 1.14
Parameter Dispersion Type:
Value:
Estimation Comments
6. Secondary Outcome
Title Phase 2: Time to Progression (TTP) Based on Tumor Assessment by Investigator
Description TTP was defined as the time in months from randomization to date of the observation of radiological PD (based on RECIST v1.1) assessed by the investigator. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm.
Time Frame From randomization to date of the observation of radiological progressive disease, assessed up to maximum 2.8 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Median (90% Confidence Interval) [Months]
5.6
2.8
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.0059
Comments
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.45
Confidence Interval () 90%
0.28 to 0.73
Parameter Dispersion Type:
Value:
Estimation Comments
7. Secondary Outcome
Title Phase 1b: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-infinity) of Tepotinib
Description The AUC(0-inf) was estimated by determining the total area under the curve of the concentration versus time curve extrapolated to infinity.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The Pharmacokinetic (PK) analysis included all participants who had received at least 1 dose of Tepotinib and who had provided at least 1 plasma concentration measurement of Tepotinib after the first dose.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1
NA
(NA)
NA
(NA)
NA
(NA)
Day 15
NA
(NA)
NA
(NA)
NA
(NA)
8. Secondary Outcome
Title Phase 1b: Area Under the Plasma Concentration-Time Curve From Time Zero to the Last Sampling Time (AUC 0-t) of Tepotinib
Description Area under the plasma concentration versus time curve from time zero to the last sampling time t at which the concentration is at or above the lower limit of quantification (LLLQ). AUC(0-t) was calculated according to the mixed log-linear trapezoidal rule.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis set was used. Here, "Overall number of participants analyzed" signifies those participants who were evaluable for this outcome measure and "Number Analyzed" signifies those participants who were evaluable at each specified time point.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 5 14 6
Day 1 of Cycle 1
4700
(12.1)
6760
(28.0)
11900
(40.3)
Day 15 of Cycle 1
11800
(35.7)
16700
(29.7)
28600
(38.8)
9. Secondary Outcome
Title Phase 1b: Area Under the Plasma Concentration-Time Curve Within 1 Dosing Interval (AUC 0-tau) of Tepotinib
Description AUC (0-tau) is the area under the plasma concentration time curve within 1 dosing interval.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis set was used. Here, "Overall number of participants analyzed" signifies those participants who were evaluable for this outcome measure and "Number Analyzed" signifies those participants who were evaluable at each specified time point.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 5 14 6
Day 1 of Cycle 1
4700
(12.1)
6760
(28.0)
11900
(40.3)
Day 15 of Cycle 1
11800
(35.7)
16700
(29.7)
28600
(38.8)
10. Secondary Outcome
Title Phase 1b: Maximum Observed Plasma Concentration (Cmax) of Tepotinib
Description Cmax is the maximum observed plasma concentration obtained directly from the concentration versus time curve.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis set was used. Here "Number Analyzed" signifies those participants who were evaluable for specified time point.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1 of Cycle 1
266
(24.7)
394
(30.4)
680
(44.0)
Day 15 of Cycle 1
585
(30.8)
815
(31.6)
1370
(36.3)
11. Secondary Outcome
Title Phase 1b: Minimum Observed Plasma Concentration (Cmin) of Tepotinib
Description Cmin is minimum observed plasma concentration obtained directly from the concentration versus time curve.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis set was used. Here, "Number of participants analyzed" signifies those participants who were evaluable for this outcome measure.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 5 11 6
Geometric Mean (Geometric Coefficient of Variation) [ng/mL]
398
(39.0)
529
(43.6)
1010
(38.8)
12. Secondary Outcome
Title Phase 1b: Average Observed Plasma Concentration (Cav) of Tepotinib
Description Cavg is the average plasma concentration within 1 dosing interval obtained directly from the concentration versus time curve.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis set was used. Here, "Number of participants analyzed" signifies those participants who were evaluable for this outcome measure.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 5 11 6
Geometric Mean (Geometric Coefficient of Variation) [ng/mL]
494
(35.7)
696
(29.7)
1190
(38.8)
13. Secondary Outcome
Title Phase 1b: Time to Reach Maximum Plasma Concentration (Tmax) of Tepotinib
Description Tmax is time to reach maximum observed plasma concentration obtained directly from the concentration versus time curve.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis set was used. Here "Number Analyzed" signifies those participants who were evaluable for specified time point.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1 of Cycle 1
8.00
8.00
10.00
Day 15 of Cycle 1
6.00
8.00
8.00
14. Secondary Outcome
Title Phase 1b: Apparent Volume of Distribution During the Terminal Phase (Vz/f) of Tepotinib
Description The Vz/f was defined as the theoretical volume in which the total amount of required to uniformly distribute to produce the desired plasma concentration. Apparent volume of distribution after oral dose (Vz/F) was influenced by the fraction absorbed. The Vz/f was calculated by dividing the dose with area under the concentration time curve from time zero to infinity multiplied with terminal elimination rate constant Lambda(z). Vz/f=Dose/AUC(0-inf)* Lambda(z).
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis included all participants who had received at least 1 dose of Tepotinib and who had provided at least 1 plasma concentration measurement of Tepotinib after the first dose.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1
NA
(NA)
NA
(NA)
NA
(NA)
Day 15
NA
(NA)
NA
(NA)
NA
(NA)
15. Secondary Outcome
Title Phase 1b: Apparent Total Body Clearance From Plasma (CL/f) of Tepotinib
Description The CL/f is a measure of the rate at which it was metabolized or eliminated by normal biological processes. Clearance obtained after oral dose was influenced by the fraction of the dose absorbed. The CL/F from plasma was calculated using the formula: Dose divided by area under the concentration time curve from time zero to infinity (AUC0-inf).
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis included all participants who had received at least 1 dose of Tepotinib and who had provided at least 1 plasma concentration measurement of Tepotinib after the first dose.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1
NA
(NA)
NA
(NA)
NA
(NA)
Day 15
NA
(NA)
NA
(NA)
NA
(NA)
16. Secondary Outcome
Title Phase 1b: Apparent Volume of Distribution During the Steady State (Vss/f) of Tepotinib
Description Volume of distribution was defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired plasma concentration of a drug. Vss/f after oral dose was influenced by the fraction absorbed.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis included all participants who had received at least 1 dose of Tepotinib and who had provided at least 1 plasma concentration measurement of Tepotinib after the first dose.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1
NA
(NA)
NA
(NA)
NA
(NA)
Day 15
NA
(NA)
NA
(NA)
NA
(NA)
17. Secondary Outcome
Title Phase 1b: Apparent Terminal Elimination Rate Constant Lambda(z) of Tepotinib
Description Lambda(z) was determined from the terminal slope of the log-transformed plasma concentration curve using linear regression method.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis included all participants who had received at least 1 dose of Tepotinib and who had provided at least 1 plasma concentration measurement of Tepotinib after the first dose.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1
NA
(NA)
NA
(NA)
NA
(NA)
Day 15
NA
(NA)
NA
(NA)
NA
(NA)
18. Secondary Outcome
Title Phase 1b: Apparent Terminal Half-life (t1/2) of Tepotinib
Description Apparent terminal half-life was defined as the time required for the plasma concentration of drug to decrease 50 percent in the final stage of its elimination.
Time Frame Predose and at 0.25, 0.5, 1, 2, 4, 8, 10, and 24 hours postdose; Day 1 and Day 15 of Cycle 1 (each Cycle is 21 days)

Outcome Measure Data

Analysis Population Description
The PK analysis included all participants who had received at least 1 dose of Tepotinib and who had provided at least 1 plasma concentration measurement of Tepotinib after the first dose.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 7 14 6
Day 1
NA
NA
NA
Day 15
NA
NA
NA
19. Secondary Outcome
Title Phase 2: Time-to-Symptomatic Progression (TTSP)
Description Time-to-symptomatic progression was defined as time (in months) from first study drug administration to the date of deterioration of symptoms assessed by Functional Assessment of Cancer Therapy Hepatobiliary Symptom Index 8 (FHSI-8) (defined as at least a 4-point increase, i.e., higher score, compared with baseline value), or deterioration to Eastern Cooperative Oncology Group (ECOG) performance score 4, or death. FHSI-8 assesses hepatobiliary cancer symptoms with total score ranges from 0 to 32 (0 = the best quality of life; 32 = the worst quality of life with severe symptoms). ECOG assess participant's performance status on a scale of 0 to 5, where 0=fully active and 5=dead.
Time Frame Up to 6.9 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment. As per planned analysis, data for this outcome was analyzed only for phase 2 based on combined analysis of both FHSI-8 and ECOG.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Median (90% Confidence Interval) [Months]
2.2
2.7
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.8915
Comments
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 1.04
Confidence Interval () 90%
0.62 to 1.77
Parameter Dispersion Type:
Value:
Estimation Comments
20. Secondary Outcome
Title Phase 2: Objective Response Rate (ORR) Based on Tumor Assessment by the IRC
Description The objective response rate (ORR) was defined as the percentage of participants who had achieved complete response (CR) or partial response (PR) as the best overall response according to radiological assessments as adjudicated by the IRC from randomization until the first occurrence of PD. CR: Complete Response (CR) defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Partial response (PR) defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started.
Time Frame Time from randomization until the first occurrence of PD assessed up to 6.9 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Number (90% Confidence Interval) [Percentage of participants]
10.5
150%
0
0%
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.0438
Comments
Method Cochran-Mantel-Haenszel
Comments
21. Secondary Outcome
Title Phase 2: Percentage of Participants With Disease Control Based on Tumor Assessment by IRC According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
Description Disease control was defined as CR, PR, or stable disease (SD) as the best overall response according to radiological assessments as adjudicated by the IRC from randomization until the first occurrence of PD. CR defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD was defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started. Percentage of participants with disease control were reported.
Time Frame Time from randomization until the first occurrence of PD assessed up to 6.9 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Number (90% Confidence Interval) [Percentage of participants]
50
714.3%
21.6
154.3%
22. Secondary Outcome
Title Phase 2: Progression-free Survival (PFS) Based on Tumor Assessment by the Investigator
Description Progression-free survival (assessed by the Investigator) time was defined as the time in months from randomization to either first observation of radiologically confirmed progression disease by the investigator or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. PFS was measured using Kaplan-Meier (KM) estimates.
Time Frame Time from randomization to disease progression or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment, assessed up to 6.9 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Median (90% Confidence Interval) [Months]
3.2
2.8
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.0496
Comments
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.59
Confidence Interval (2-Sided) 90%
0.38 to 0.92
Parameter Dispersion Type:
Value:
Estimation Comments
23. Secondary Outcome
Title Phase 2: Objective Response Rate (ORR) Based on Tumor Assessment by the Investigator
Description The objective response rate was defined as the percentage of participants who had achieved CR or PR as the best overall response according to radiological assessments as adjudicated by the investigator from randomization until the first occurrence of PD. CR defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD was defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started.
Time Frame Time from randomization until the first occurrence of PD assessed up to 6.9 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Number (90% Confidence Interval) [Percentage of participants]
15.8
225.7%
2.7
19.3%
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection Phase 1b: Tepotinib 300 mg, Phase 1b: Tepotinib 500 mg
Comments
Type of Statistical Test Equivalence
Comments Sample size required 100 TTP events to ensure 80% power with a two-sided significance level of 10% for rejecting the null hypothesis of equal treatment effect between treatment arms.
Statistical Test of Hypothesis p-Value 0.0527
Comments
Method Cochran-Mantel-Haenszel
Comments
24. Secondary Outcome
Title Phase 2: Percentage of Participants With Disease Control Based on Tumor Assessment by Investigator According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
Description Disease control was defined as CR, PR, or stable disease (SD) as the best overall response according to radiological assessments as adjudicated by the IRC from randomization until the first occurrence of PD. CR defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD was defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started. Percentage of participants with disease control were reported.
Time Frame Approximately up to 6.9 years

Outcome Measure Data

Analysis Population Description
The mITT analysis set in the Phase 2 part of this study included all participants with MET+ HCC who were randomized to study treatment.
Arm/Group Title Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
Measure Participants 38 37
Number (90% Confidence Interval) [Percentage of Participants]
60.5
864.3%
45.9
327.9%

Adverse Events

Time Frame Baseline up to Day 30 after the last dose of study treatment, assessed up to 94 weeks (Phase 1b) and 6.9 years (Phase 2).
Adverse Event Reporting Description The safety analysis set included all participants who had received any dose of the study medication.
Arm/Group Title Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg Phase 2: Tepotinib Phase 2 Sorafenib
Arm/Group Description Participants received Tepotinib 300 milligram (mg) orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 500 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants received Tepotinib 1000 mg orally once daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Tepotinib recommended Phase 2 dose (RP2D) determined from Phase 1b over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal. Participants randomized to receive Sorafenib 400 mg orally twice daily over a 21-day cycle until disease progression, intolerable toxicity or participant withdrawal.
All Cause Mortality
Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg Phase 2: Tepotinib Phase 2 Sorafenib
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 7/7 (100%) 11/14 (78.6%) 3/6 (50%) 24/45 (53.3%) 30/44 (68.2%)
Serious Adverse Events
Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg Phase 2: Tepotinib Phase 2 Sorafenib
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 2/7 (28.6%) 9/14 (64.3%) 4/6 (66.7%) 23/45 (51.1%) 12/44 (27.3%)
Blood and lymphatic system disorders
Thrombocytopenia 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Cardiac disorders
Supraventricular tachycardia 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Gastrointestinal disorders
Abdominal pain 0/7 (0%) 1/14 (7.1%) 1/6 (16.7%) 2/45 (4.4%) 2/44 (4.5%)
Ascites 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 2/45 (4.4%) 0/44 (0%)
Diarrhoea 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 1/45 (2.2%) 1/44 (2.3%)
Duodenal ulcer 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Encapsulating peritoneal sclerosis 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Ileus 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Melaena 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Upper gastrointestinal haemorrhage 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 1/45 (2.2%) 1/44 (2.3%)
Duodenal ulcer haemorrhage 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Peritoneal haemorrhage 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
General disorders
Disease progression 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 7/45 (15.6%) 2/44 (4.5%)
Asthenia 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 1/44 (2.3%)
Hernia 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Oedema peripheral 0/7 (0%) 0/14 (0%) 0/6 (0%) 2/45 (4.4%) 0/44 (0%)
Hepatobiliary disorders
Cholecystitis acute 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 1/44 (2.3%)
Hepatic failure 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 1/44 (2.3%)
Hepatic function abnormal 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Infections and infestations
Candida infection 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Cellulitis 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Enteritis infectious 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Pneumonia 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Investigations
Alanine aminotransferase increased 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Aspartate aminotransferase increased 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Blood bilirubin increased 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 1/44 (2.3%)
Lipase increased 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 1/44 (2.3%)
Metabolism and nutrition disorders
Decreased appetite 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 1/45 (2.2%) 0/44 (0%)
Hyperglycaemia 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Musculoskeletal and connective tissue disorders
Muscular weakness 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Musculoskeletal pain 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Gouty arthritis 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Metastases to central nervous system 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 1/44 (2.3%)
Bladder cancer 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Nervous system disorders
Cerebrovascular accident 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Haemorrhage intracranial 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 1/44 (2.3%)
Hepatic encephalopathy 0/7 (0%) 0/14 (0%) 0/6 (0%) 2/45 (4.4%) 1/44 (2.3%)
Paraesthesia 0/7 (0%) 0/14 (0%) 0/6 (0%) 2/45 (4.4%) 0/44 (0%)
Seizure 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Respiratory, thoracic and mediastinal disorders
Dyspnoea exertional 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Haemoptysis 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Pleural effusion 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Skin and subcutaneous tissue disorders
Rash maculo-papular 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Vascular disorders
Deep vein thrombosis 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 1/44 (2.3%)
Hypertension 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Lymphoedema 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Other (Not Including Serious) Adverse Events
Phase 1b: Tepotinib 300 mg Phase 1b: Tepotinib 500 mg Phase 1b: Tepotinib 1000 mg Phase 2: Tepotinib Phase 2 Sorafenib
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 7/7 (100%) 14/14 (100%) 6/6 (100%) 43/45 (95.6%) 43/44 (97.7%)
Blood and lymphatic system disorders
Anaemia 0/7 (0%) 2/14 (14.3%) 1/6 (16.7%) 4/45 (8.9%) 2/44 (4.5%)
Thrombocytopenia 0/7 (0%) 0/14 (0%) 0/6 (0%) 3/45 (6.7%) 2/44 (4.5%)
Eye disorders
Dry eye 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Gastrointestinal disorders
Abdominal discomfort 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Abdominal distension 4/7 (57.1%) 4/14 (28.6%) 0/6 (0%) 9/45 (20%) 5/44 (11.4%)
Abdominal mass 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Abdominal pain 2/7 (28.6%) 6/14 (42.9%) 1/6 (16.7%) 11/45 (24.4%) 9/44 (20.5%)
Abdominal pain upper 1/7 (14.3%) 2/14 (14.3%) 1/6 (16.7%) 2/45 (4.4%) 7/44 (15.9%)
Anorectal discomfort 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Ascites 0/7 (0%) 3/14 (21.4%) 3/6 (50%) 11/45 (24.4%) 4/44 (9.1%)
Constipation 1/7 (14.3%) 4/14 (28.6%) 4/6 (66.7%) 6/45 (13.3%) 8/44 (18.2%)
Diarrhoea 2/7 (28.6%) 6/14 (42.9%) 4/6 (66.7%) 16/45 (35.6%) 17/44 (38.6%)
Dyspepsia 0/7 (0%) 3/14 (21.4%) 1/6 (16.7%) 4/45 (8.9%) 6/44 (13.6%)
Gastric ulcer 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Gingival bleeding 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 4/44 (9.1%)
Gastritis 0/7 (0%) 1/14 (7.1%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Nausea 3/7 (42.9%) 3/14 (21.4%) 2/6 (33.3%) 4/45 (8.9%) 7/44 (15.9%)
Stomatitis 1/7 (14.3%) 1/14 (7.1%) 0/6 (0%) 1/45 (2.2%) 3/44 (6.8%)
Vomiting 1/7 (14.3%) 3/14 (21.4%) 1/6 (16.7%) 3/45 (6.7%) 6/44 (13.6%)
General disorders
Asthenia 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 5/44 (11.4%)
Chest pain 0/7 (0%) 0/14 (0%) 0/6 (0%) 3/45 (6.7%) 2/44 (4.5%)
Fatigue 1/7 (14.3%) 3/14 (21.4%) 2/6 (33.3%) 12/45 (26.7%) 13/44 (29.5%)
Disease progression 0/7 (0%) 0/14 (0%) 0/6 (0%) 3/45 (6.7%) 1/44 (2.3%)
Influenza like illness 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Localised oedema 1/7 (14.3%) 1/14 (7.1%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Malaise 1/7 (14.3%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Non-cardiac chest pain 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Oedema 0/7 (0%) 0/14 (0%) 0/6 (0%) 3/45 (6.7%) 1/44 (2.3%)
Oedema peripheral 2/7 (28.6%) 4/14 (28.6%) 1/6 (16.7%) 20/45 (44.4%) 1/44 (2.3%)
Pain 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Pyrexia 0/7 (0%) 4/14 (28.6%) 2/6 (33.3%) 7/45 (15.6%) 7/44 (15.9%)
Hepatobiliary disorders
Hepatorenal syndrome 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Immune system disorders
Anaphylactic reaction 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Infections and infestations
Rash pustular 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 3/44 (6.8%)
Upper respiratory tract infection 0/7 (0%) 2/14 (14.3%) 0/6 (0%) 3/45 (6.7%) 3/44 (6.8%)
Bacteraemia 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Cellulitis 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Infection 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Investigations
Alanine aminotransferase increased 2/7 (28.6%) 1/14 (7.1%) 3/6 (50%) 7/45 (15.6%) 9/44 (20.5%)
Amylase increased 0/7 (0%) 2/14 (14.3%) 1/6 (16.7%) 4/45 (8.9%) 7/44 (15.9%)
Aspartate aminotransferase increased 3/7 (42.9%) 3/14 (21.4%) 4/6 (66.7%) 10/45 (22.2%) 16/44 (36.4%)
Blood alkaline phosphatase increased 0/7 (0%) 0/14 (0%) 3/6 (50%) 5/45 (11.1%) 3/44 (6.8%)
Blood bilirubin increased 0/7 (0%) 1/14 (7.1%) 1/6 (16.7%) 4/45 (8.9%) 11/44 (25%)
Blood creatinine increased 1/7 (14.3%) 0/14 (0%) 3/6 (50%) 7/45 (15.6%) 1/44 (2.3%)
Creatinine renal clearance decreased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Creatinine renal clearance increased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Electrocardiogram QT prolonged 0/7 (0%) 0/14 (0%) 0/6 (0%) 3/45 (6.7%) 1/44 (2.3%)
Gamma-glutamyltransferase increased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 3/45 (6.7%) 8/44 (18.2%)
Lipase increased 0/7 (0%) 3/14 (21.4%) 1/6 (16.7%) 2/45 (4.4%) 8/44 (18.2%)
Lymphocyte count decreased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Neutrophil count decreased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 2/45 (4.4%) 4/44 (9.1%)
Platelet count decreased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 7/45 (15.6%) 7/44 (15.9%)
White blood cell count decreased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 3/44 (6.8%)
White blood cell count increased 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Weight decreased 0/7 (0%) 0/14 (0%) 0/6 (0%) 2/45 (4.4%) 9/44 (20.5%)
Metabolism and nutrition disorders
Decreased appetite 1/7 (14.3%) 3/14 (21.4%) 2/6 (33.3%) 16/45 (35.6%) 18/44 (40.9%)
Hyperglycaemia 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Hyperkalaemia 0/7 (0%) 2/14 (14.3%) 0/6 (0%) 4/45 (8.9%) 2/44 (4.5%)
Hyperuricaemia 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Hypocalcaemia 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 1/45 (2.2%) 0/44 (0%)
Hypoglycaemia 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Hyponatraemia 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 5/45 (11.1%) 2/44 (4.5%)
Hypophosphataemia 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 3/44 (6.8%)
Hypoalbuminaemia 1/7 (14.3%) 1/14 (7.1%) 4/6 (66.7%) 12/45 (26.7%) 4/44 (9.1%)
Hypokalaemia 0/7 (0%) 1/14 (7.1%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Musculoskeletal and connective tissue disorders
Back pain 0/7 (0%) 0/14 (0%) 0/6 (0%) 2/45 (4.4%) 3/44 (6.8%)
Musculoskeletal pain 1/7 (14.3%) 1/14 (7.1%) 0/6 (0%) 1/45 (2.2%) 3/44 (6.8%)
Pain in extremity 0/7 (0%) 0/14 (0%) 0/6 (0%) 4/45 (8.9%) 5/44 (11.4%)
Arthralgia 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Muscle spasms 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Muscular weakness 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Musculoskeletal chest pain 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Myalgia 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Nervous system disorders
Brain injury 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Dizziness 0/7 (0%) 2/14 (14.3%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Generalised tonic-clonic seizure 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Headache 0/7 (0%) 3/14 (21.4%) 1/6 (16.7%) 2/45 (4.4%) 5/44 (11.4%)
Metabolic encephalopathy 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Posterior reversible encephalopathy syndrome 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Tremor 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Psychiatric disorders
Insomnia 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 1/45 (2.2%) 3/44 (6.8%)
Mental status changes 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Renal and urinary disorders
Acute kidney injury 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Dysuria 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Pollakiuria 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Proteinuria 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Reproductive system and breast disorders
Penile swelling 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Scrotal swelling 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Respiratory, thoracic and mediastinal disorders
Cough 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 3/44 (6.8%)
Dysphonia 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 2/45 (4.4%) 4/44 (9.1%)
Dyspnoea 1/7 (14.3%) 1/14 (7.1%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Dyspnoea exertional 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Hiccups 1/7 (14.3%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Oropharyngeal pain 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Pleural effusion 0/7 (0%) 1/14 (7.1%) 2/6 (33.3%) 0/45 (0%) 0/44 (0%)
Pulmonary oedema 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Tachypnoea 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Skin and subcutaneous tissue disorders
Alopecia 0/7 (0%) 0/14 (0%) 0/6 (0%) 1/45 (2.2%) 11/44 (25%)
Dermatitis acneiform 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 5/44 (11.4%)
Nail disorder 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Palmar-plantar erythrodysaesthesia syndrome 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 11/45 (24.4%) 27/44 (61.4%)
Pruritus 1/7 (14.3%) 3/14 (21.4%) 2/6 (33.3%) 0/45 (0%) 0/44 (0%)
Rash 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 4/45 (8.9%) 4/44 (9.1%)
Rash generalised 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Rash maculo-papular 0/7 (0%) 2/14 (14.3%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)
Skin exfoliation 0/7 (0%) 1/14 (7.1%) 0/6 (0%) 0/45 (0%) 0/44 (0%)
Pain of skin 0/7 (0%) 0/14 (0%) 0/6 (0%) 0/45 (0%) 3/44 (6.8%)
Vascular disorders
Hypertension 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 12/44 (27.3%)
Hypotension 0/7 (0%) 1/14 (7.1%) 1/6 (16.7%) 1/45 (2.2%) 0/44 (0%)
Phlebitis 0/7 (0%) 0/14 (0%) 1/6 (16.7%) 0/45 (0%) 0/44 (0%)

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 Communication Center
Organization Merck KGaA Darmstadt, Germany
Phone +49-6151-72-5200
Email service@emdgroup.com
Responsible Party:
Merck KGaA, Darmstadt, Germany
ClinicalTrials.gov Identifier:
NCT01988493
Other Study ID Numbers:
  • 200095-004
First Posted:
Nov 20, 2013
Last Update Posted:
Mar 7, 2022
Last Verified:
Dec 1, 2021