Study Evaluating SKI-606 (Bosutinib) In Advanced Malignant Solid Tumors

January 3, 2013 updated by: Pfizer

Phase I Dose-Escalation Study Of Oral SKI-606 In Subjects With Advanced Malignant Solid Tumors

To evaluate the safety and tolerability of oral SKI-606 (bosutinib) administered on a daily schedule to subjects with advanced malignant solid tumors and to define a maximum tolerated dose (MTD) in this subject population.

Study Overview

Status

Completed

Conditions

Intervention / Treatment

Study Type

Interventional

Enrollment (Actual)

151

Phase

  • Phase 1

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

    • Alabama
      • Birmington, Alabama, United States, 35233
        • Pfizer Investigational Site
    • Arizona
      • Scottsdale, Arizona, United States, 85258
        • Pfizer Investigational Site
    • California
      • Los Angeles, California, United States, 90033
        • Pfizer Investigational Site
    • Florida
      • Tampa, Florida, United States, 33612
        • Pfizer Investigational Site
    • Georgia
      • Atlanta, Georgia, United States, 30341
        • Pfizer Investigational Site
    • Indiana
      • Indianpolis, Indiana, United States, 46202
        • Pfizer Investigational Site
    • Maryland
      • Baltimore, Maryland, United States, 21287
        • Pfizer Investigational Site
    • Michigan
      • Detroit, Michigan, United States, 48202
        • Pfizer Investigational Site
      • Lansing, Michigan, United States, 48910
        • Pfizer Investigational Site
    • New York
      • New York, New York, United States, 10032
        • Pfizer Investigational Site
      • New York, New York, United States, 10016
        • Pfizer Investigational Site
    • North Carolina
      • Charlotte, North Carolina, United States, 28203
        • Pfizer Investigational Site
      • Charlotte, North Carolina, United States, 28211
        • Pfizer Investigational Site
      • UNC Chapel Hill, North Carolina, United States, 27514
        • Pfizer Investigational Site
      • UNC Chapel Hill, North Carolina, United States, 27759
        • Pfizer Investigational Site
    • Ohio
      • Cleveland, Ohio, United States, 44106-1736
        • Pfizer Investigational Site
    • Texas
      • San Antonio, Texas, United States, 78258
        • Pfizer Investigational Site
      • Tyler, Texas, United States, 75702
        • Pfizer Investigational Site
    • Washington
      • Seattle, Washington, United States, 98104
        • Pfizer Investigational Site

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria:

  • Advanced or recurrent solid malignancy confirmed histologically or cytologically for which no effective therapy is available.
  • Eastern Cooperative Oncology Group (ECOG) performance status of 0 - 1.
  • Measurable disease as outlined by the modified Response Evaluation Criteria in Solid Tumors (RECIST) criteria.
  • Other inclusion applies.

Exclusion Criteria:

  • Use of any systemic antitumor agents or any investigational agent within 28 days before the first dose of test article is administered.
  • Prior exposure to SKI-606 or any other Src-kinase inhibitor, major surgery or radiotherapy within 14 days before the first dose of test article (recovery from previous surgery should be complete before day 1).
  • Active central nervous system (CNS) metastases, as indicated by clinical symptoms, cerebral edema, requirement for corticosteroids and/or progressive growth (Treated CNS metastases must be stable for >= 2 weeks before day 1).
  • Other exclusion applies.

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Non-Randomized
  • Interventional Model: Single Group Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Dose escalation
Dose finding study of monotherapy bosutinib in patients with advanced solid tumors.
Dose levels evaluated 50mg, 100mg, 200mg, 300mg, 400mg, 500mg and 600mg. 500mg was identified as MTD, however due to GI toxicities at that dose, 400mg was selected as the RP2D. Drug was administered as long as tolerable and disease under study did not worsen.
400mg QD bosutinib, as long as tolerated and disease under study does not worsen.
Experimental: Colorectal Cancer
Enroll 30 patients at RP2D to further evaluate safety and efficacy in subgroup population.
Dose levels evaluated 50mg, 100mg, 200mg, 300mg, 400mg, 500mg and 600mg. 500mg was identified as MTD, however due to GI toxicities at that dose, 400mg was selected as the RP2D. Drug was administered as long as tolerable and disease under study did not worsen.
400mg QD bosutinib, as long as tolerated and disease under study does not worsen.
Experimental: Pancreatic Cancer
Enroll 30 patients at RP2D to further evaluate safety and efficacy in subgroup population.
Dose levels evaluated 50mg, 100mg, 200mg, 300mg, 400mg, 500mg and 600mg. 500mg was identified as MTD, however due to GI toxicities at that dose, 400mg was selected as the RP2D. Drug was administered as long as tolerable and disease under study did not worsen.
400mg QD bosutinib, as long as tolerated and disease under study does not worsen.
Experimental: Non-Small Cell Lung Cancer (NSCLC)
Enroll 30 patients at RP2D to further evaluate safety and efficacy in subgroup population.
Dose levels evaluated 50mg, 100mg, 200mg, 300mg, 400mg, 500mg and 600mg. 500mg was identified as MTD, however due to GI toxicities at that dose, 400mg was selected as the RP2D. Drug was administered as long as tolerable and disease under study did not worsen.
400mg QD bosutinib, as long as tolerated and disease under study does not worsen.

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Number of Participants With Dose-limiting Toxicities (DLT) in Part 1
Time Frame: Part 1 Baseline up to Day 28
DLT included any grade 3 or 4 clinically-evident non-hematologic toxicity, grade 4 neutropenia of greater than or equal to (>=) 7-day duration or with fever >= 38.5 degrees Celsius (febrile neutropenia); grade 4 thrombocytopenia >= 2-day duration or with bleeding requiring platelet transfusion, any clinically-significant grade >= 2 toxicity that requires >=14 days to resolve (to less than or equal to [=<] grade 1) which occurred in first 21 days of study and considered at least possibly related to bosutinib.
Part 1 Baseline up to Day 28
Number of Participants With Adverse Events (AEs) by Seriousness
Time Frame: Baseline up to 30 days after last dose
Counts of participants who had treatment-emergent adverse events (TEAEs), defined as newly occurring or worsening after first dose. Participants with multiple occurrences of an AE within a category were counted once within the category.
Baseline up to 30 days after last dose
Duration of Most Frequently Observed Adverse Events (AEs)
Time Frame: Baseline up to 30 days after last dose
The most frequently observed treatment-emergent AEs were gastrointestinal disorders which included diarrhea, nausea and vomiting. Duration of AE per event is calculated as AE stop date minus AE start date plus 1.
Baseline up to 30 days after last dose
Number of Participants With Best Overall Response (BOR) in Part 1
Time Frame: Part 1 Baseline, last week (Day 15 to 23) of cycles 2, 4, 6, 8 and thereafter every 3 cycles up to 30 days after last dose
BOR:investigator assessment by modified Response Evaluation Criteria in Solid Tumors (RECIST), recorded from treatment start until disease progression/recurrence. Complete Response:disappearance of all lesions. Partial Response (PR):>=30% decrease in sum of longest diameters (SLDs) of target lesions (TLs) taking as reference baseline SLD. Progressive disease (PD):>=20% increase in SLD of TLs taking as reference smallest SLD since treatment start, or appearance of >=1 new lesion. Stable disease: neither shrinkage for PR nor increase for PD taking as reference smallest SLD since treatment start.
Part 1 Baseline, last week (Day 15 to 23) of cycles 2, 4, 6, 8 and thereafter every 3 cycles up to 30 days after last dose
Number of Participants With Best Overall Response (BOR) in Part 2
Time Frame: Part 2 Baseline, last week (Day 15 to 23) of cycles 2, 4, 6, 8 and thereafter every 3 cycles up to 30 days after last dose
BOR: investigator assessment by modified RECIST, recorded from treatment start until disease progression/recurrence. Complete Response: disappearance of all lesions. PR: >=30% decrease in SLDs of TLs taking as reference baseline SLD. PD: >=20% increase in SLD of TLs taking as reference smallest SLD since treatment start, or appearance of >=1 new lesion. Stable disease: neither shrinkage for PR nor increase for PD taking as reference smallest SLD since treatment start.
Part 2 Baseline, last week (Day 15 to 23) of cycles 2, 4, 6, 8 and thereafter every 3 cycles up to 30 days after last dose
Maximum Tolerated Dose (MTD) in Part 1
Time Frame: Part 1 Day 1 up to Day 28
MTD: highest dose level at which not more than 1 of 6 participants experienced DLT after 21 days of treatment (Cycle 1). DLT included any grade 3 or 4 clinically-evident non-hematologic toxicity, grade 4 neutropenia of >= 7-day duration or with fever >= 38.5 degrees Celsius (febrile neutropenia); grade 4 thrombocytopenia >= 2-day duration or with bleeding requiring platelet transfusion, any clinically-significant grade >= 2 toxicity that requires >=14 days to resolve (to =< grade 1) which occurred in first 21 days of study and considered at least possibly related to bosutinib.
Part 1 Day 1 up to Day 28

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Maximum Tolerated Dose (MTD) for Prolonged Use
Time Frame: Part 1 Day 1 up to Day 28
MTD for prolonged use was the highest dose level at which not more than 1 of 6 participants experienced DLT after 21 days of treatment (Cycle 1) and was selected as recommended dose in Phase 2, due to substantial number of Grade 2 gastrointestinal toxicities observed in the MTD lead-in cohort (500 mg).
Part 1 Day 1 up to Day 28
Number of Participants With Change From Baseline in Laboratory Test Results
Time Frame: Baseline up to end of treatment (Week 95)
Criteria for potentially clinically significant (PCS) laboratory values: albumin <20, hemoglobin <80 gram/liter(g/L); alkaline phosphatase, aspartate aminotransferase, alanine aminotransferase >5*upper limit of normal(ULN) milliunit/milliliter(mU/mL); bilirubin total, creatinine>3*ULN micromole/L; calcium <1.75 and >3.1,potassium <3 and >6, sodium <130, glucose <2.2,phosphorous <0.6 millimole/L; international normalized ratio >2*ULN, partial thromboplastin time, prothrombin time >2*ULN seconds; platelet count <50*10^9/L. Participants meeting at least 1 PCS criteria are reported.
Baseline up to end of treatment (Week 95)
Number of Participants With Change From Baseline in Electrocardiogram (ECG) and Chest X-ray
Time Frame: Baseline up to end of treatment (Week 95)
Number of participants with PCS ECG findings is reported on-therapy (OT) and at final visit (FV). Criteria for PCS ECG findings: heart rate (HR) =<45 beats/minute (bpm) and decrease (Dec) >15/>=120 bpm and decrease of >15 bpm; PR interval (Int) >=220 millisecond (msec), increase (Inc) >=20 msec, QRS Int >=120 msec, corrected QT (QTc) and QTc using fridericia formula(QTcF) Int >500 msec, increase >60 msec; no sinus rhythm; overall ECG abnormal. Participants with at least 1 measurement exceeding the criteria for PCS are reported.
Baseline up to end of treatment (Week 95)
Concomitant Medications Used for Management of Adverse Events (AEs)
Time Frame: Day 1 up to end of treatment (Week 95)
Number of participants taking any non-study medications which were administered from Day 1 up to end of treatment (Week 95) as a management of an AE was to be reported.
Day 1 up to end of treatment (Week 95)
Change From Baseline in Karnofsky Performance Score
Time Frame: Baseline up to end of treatment (Week 95)
Karnofsky performance score is used to quantify participant's general well-being and activities of daily life and participants are classified based on their functional impairment. Karnofsky performance score is 11 level score which ranges between 0 (death) to 100 (complete healthy status). Higher score means higher ability to perform daily tasks.
Baseline up to end of treatment (Week 95)
Number of Participants With Change From Baseline in Physical Examination
Time Frame: Baseline up to end of treatment (Week 95)
Physical examinations included body weight, height and vital signs and only finding that exceeded the criterion for PCS was weight. Criteria for weight was: an increase or decrease of >=10% from baseline.
Baseline up to end of treatment (Week 95)
Number of Participants With Change From Baseline in Opthalmologic Examination
Time Frame: Baseline up to end of treatment (Week 95)
Ophthalmologic evaluation included visual acuity, funduscopic examination, and any clinically-significant abnormality.
Baseline up to end of treatment (Week 95)
Overall Survival (OS) in Part 2
Time Frame: Part 2 Baseline until death or 3, 6, 9 and 12 months after treatment discontinuation
Time in weeks from the start of study treatment to date of death due to any cause. OS was calculated as (the death date minus the date of first dose of study medication plus 1) divided by 7. Death was determined from death case report forms (CRFs) or from follow-up contact data (where the participant current status was death).
Part 2 Baseline until death or 3, 6, 9 and 12 months after treatment discontinuation
Progression Free Survival (PFS) in Part 2
Time Frame: Part 2 Baseline until death or 3, 6, 9 and 12 months after treatment discontinuation
Time in weeks from start of study treatment to first documentation of objective tumor progression or death due to any cause. PFS was calculated as (first event date minus the date of first dose of study medication plus 1) divided by 7. Tumor progression was determined from oncologic assessment data (where data meet the criteria for PD, or from death CRFs).
Part 2 Baseline until death or 3, 6, 9 and 12 months after treatment discontinuation
Maximum Observed Plasma Concentration (Cmax)
Time Frame: 0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14
0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14
Time to Reach Maximum Observed Plasma Concentration (Tmax)
Time Frame: 0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14
0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14
Plasma Decay Half-Life (t1/2)
Time Frame: 0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14
Plasma decay half-life is the time measured for the plasma concentration to decrease by one half.
0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14
Area Under the Concentration-Time Curve (AUC)
Time Frame: 0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14
AUC is a measure of the serum concentration of the drug over time. It is used to characterize drug absorption. Steady state concentration was achieved at Day 15.
0 hour (pre-dose) on Day 1, 0 (pre-dose), 1, 2, 3, 4, 6, 8, 24 (0 hour [pre-dose] on Day 15) hours post-dose on Day 14

Other Outcome Measures

Outcome Measure
Measure Description
Time Frame
Gene Expression at Baseline
Time Frame: Baseline
Gene expression profile was evaluated by measuring transcript levels of messenger RNA (mRNA) in peripheral blood samples. Expression profiling of mRNA: done to measure the expressed genome of mRNA transcripts or done in a gene-specific targeted manner.
Baseline

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Sponsor

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start

October 1, 2004

Primary Completion (Actual)

November 1, 2007

Study Completion (Actual)

November 1, 2007

Study Registration Dates

First Submitted

September 12, 2005

First Submitted That Met QC Criteria

September 12, 2005

First Posted (Estimate)

September 19, 2005

Study Record Updates

Last Update Posted (Estimate)

February 11, 2013

Last Update Submitted That Met QC Criteria

January 3, 2013

Last Verified

January 1, 2013

More Information

Terms related to this study

Keywords

Other Study ID Numbers

  • 3160A1-100
  • B1871012

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Neoplasms

Clinical Trials on bosutinib

3
Subscribe