A Study Of Combined C- MET Inhibitor And PAN-HER Inhibitor (PF-02341066 And PF-00299804) In Patients With Non- Small Cell Lung Cancer

Sponsor
Pfizer (Industry)
Overall Status
Completed
CT.gov ID
NCT01121575
Collaborator
(none)
70
10
2
42.1
7
0.2

Study Details

Study Description

Brief Summary

Lung cancer tumors become resistant to the first generation epidermal growth factor receptor (EGFR) inhibitors erlotinib or gefitinib by changing and increasing the activity of two cell signaling pathways: the cMET pathway and the EGFR pathway. Both resistance mechanisms can occur at the same time, in the same patient and even in the same tumor. This study combines a second generation EGFR inhibitor and a cMET inhibitor to block both these pathways in order to overcome resistance and treat this disease.

Condition or Disease Intervention/Treatment Phase
Phase 1

Study Design

Study Type:
Interventional
Actual Enrollment :
70 participants
Allocation:
Non-Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
A Phase 1, Open-label, Dose Escalation Study To Evaluate Safety, Pharmacokinetics And Pharmacodynamics Of Combined Oral C-met/Alk Inhibitor (Pf-02341066) And Pan-her Inhibitor (Pf-00299804) In Patients With Advanced Non-small Cell Lung Cancer
Study Start Date :
Aug 1, 2010
Actual Primary Completion Date :
Feb 1, 2014
Actual Study Completion Date :
Feb 1, 2014

Arms and Interventions

Arm Intervention/Treatment
Experimental: Arm 1

PF-02341066 AND PF-00299804: Patients will be treated with combined cMET inhibitor (PF-02341066) and panHER inhibitor (PF-00299804).

Drug: PF-02341066
Arm 1: The starting dose will be 200 mg by mouth, twice a day of PF 02341066 in tablet form The dose of each drug in the combination [PF-02341066 and PF-00299804] will be escalated or de-escalated until the maximum tolerated combined dose is reached. Patients will then be treated with the maximum tolerated combined dose.
Other Names:
  • cMET inhibitor
  • Drug: PF-00299804
    Arm 1: The starting dose will be 30 mg by mouth once a day of PF-0029804 in tablet form. The dose of each drug in the combination [PF-02341066 and PF-00299804] will be escalated or de-escalated until the maximum tolerated combined dose is reached. Patients will then be treated with the maximum tolerated combined dose.
    Other Names:
  • panHER inhibitor
  • Experimental: Arm 2

    PF-00299804 FOLLOWED BY COMBINED PF-02341066 AND PF-00299804: Patients will be treated with single agent panHER inhibitor (PF-00299804) until disease progression and then with the maximum tolerated combined dose of cMET inhibitor (PF-02341066) and panHER inhibitor (PF-00299804).

    Drug: PF-02341066
    Arm 2: With progressive disease the maximum tolerated combined dose of PF-02341066 (given by mouth twice a day in tablet form) and PF-00299804 (given by mouth once a day in tablet form).
    Other Names:
  • cMET inhibitor
  • Drug: PF-00299804
    Arm 2: The dose of 45 mg by mouth once a day of PF-00299804 in tablet form until progressive disease and then the maximum tolerated combined dose of PF-02341066 (given by mouth twice a day in tablet form) and PF-00299804 (given by mouth once a day in tablet form).
    Other Names:
  • panHER inhibitor
  • Outcome Measures

    Primary Outcome Measures

    1. Overview of Treatment-emergent All Causalities Adverse Events (AEs) in Escalation Phase [Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)]

      AE was any untoward medical occurrence with study drug/ device in a trial participant. Serious adverse event (SAE) was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using National Cancer Institute (NCI) Common Terminology Criteria for AEs (CTCAE) v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.

    2. Overview of Treatment-emergent All Causalities AEs in Expansion Phase [Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)]

      AE was any untoward medical occurrence with study drug/ device in a trial participant. SAE was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using NCI CTCAE v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.

    3. Overview of Treatment-emergent, Treatment-related AEs in Escalation Phase [Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)]

      An AE was any untoward medical occurrence attributed to study treatment in a participant who received study treatment. SAE was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using NCI CTCAE v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.

    4. Overview of Treatment-emergent, Treatment-related AEs in Expansion Phase [Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)]

      An AE was any untoward medical occurrence attributed to study treatment in a participant who received study treatment. SAE was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using NCI CTCAE v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.

    5. Number of Participants With Dose Limiting Toxicities (DLTs) in Escalation Phase [Cycle 1 (4 weeks)]

      DLTs were those AEs which occurred in Cycle 1 of treatment in Dose Escalation Phase which may be attributed to study drug [combined Crizotinib (PF-02341066) plus Dacomitinib (PF-00299804)] without a clear alternative explanation and despite the use of adequate/maximal medical intervention as dictated by local institutional clinical practices or the judgment of the investigator. The following events were considered DLTs (using CTCAE version 4.02);1. Grade ≥4 hematologic events. 2. Grade ≥3 non-hematological events (except Grade 3/4 asymptomatic hypophosphatemia and Grade 3/4 hyperuricemia without signs and symptoms of gout). Nausea, vomiting or diarrhea had to have persisted at Grade 3 or 4 despite maximal medical therapy. Grade 3 hypertension will be considered a DLT only if the event is unmanageable by standard approved pharmacologic agents or if the symptomatic sequelae are identified despite appropriate medical intervention.

    Secondary Outcome Measures

    1. Number of Participants With Stable Disease and Stable Disease Duration in Escalation Phase [From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)]

      If a participant had not achieved an objective response with confirmed complete response (CR) or partial response (PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) - 1.1 as determined by the investigators, relative to the response evaluable population, but remained stable for at least 6 weeks after first dose, then the best overall response for such a participant was considered as stable disease.

    2. Number of Participants With Stable Disease and Stable Disease Duration in Expansion Phase [From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)]

      If a participant had not achieved an objective response with confirmed complete response (CR) or partial response (PR) according to RECIST (1.1) as determined by the investigators, relative to the response evaluable population, but remained stable for at least 6 weeks after first dose, then the best overall response for such a participant was considered as stable disease.

    3. Number of Participants With Objective Response Rate (ORR) in Escalation Phase [From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)]

      ORR was defined as the percent of participants with CR or PR according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1) determined by study physicians, relative to the response evaluable population. Participants were considered non-responders until proven otherwise. Thus, participants who: 1) Did not have CR or PR while on study, or 2) Did not have a post-baseline tumor evaluation, or 3) Received anti-tumor treatment other than the study medication prior to reaching a CR or PR, or 4) Died, in progress, or drop out for any reason prior to reaching a CR or PR, were counted as non-responders in the assessment of ORR. To be assigned a status of PR or CR, changes in tumor measurements in participants with responding tumors were confirmed by repeated tumor assessment that were performed 4 weeks after the criteria for response were first met.

    4. Number of Participants With ORR in Expansion Phase [From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)]

      ORR was defined as the percent of participants with CR or PR according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1) determined by study physicians, relative to the response evaluable population. Participants were considered non-responders until proven otherwise. Thus, participants who: 1) Did not have CR or PR while on study, or 2) Did not have a post-baseline tumor evaluation, or 3) Received anti-tumor treatment other than the study medication prior to reaching a CR or PR, or 4) Died, in progress, or drop out for any reason prior to reaching a CR or PR, were counted as non-responders in the assessment of ORR. To be assigned a status of PR or CR, changes in tumor measurements in participants with responding tumors were confirmed by repeated tumor assessment that were performed 4 weeks after the criteria for response were first met.

    5. Duration of Response for the Only Participant Shown Partial Response in Expansion Phase [From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)]

      This outcome measure presented the duration of response for one participant in expansion cohort 1 who showed partial response.

    6. Progression Free Survival (PFS) in Escalation Phase [From randomization to objective tumor progression or death (up to post treatment follow-up as 28-35 days after last dose of study treatment)]

      PFS was defined as the time from the date of the first dose to the date of the first documentation of objective tumor progression according to RECIST 1.1 or death on study due to any cause, whichever occurred first. If tumor progression data included more than 1 date, the first date was used.

    7. Progression Free Survival (PFS) in Expansion Phase [From randomization to objective tumor progression or death (up to post treatment follow-up as 28-35 days after last dose of study treatment)]

      PFS was defined as the time from the date of the first dose to the date of the first documentation of objective tumor progression according to RECIST 1.1 or death on study due to any cause, whichever occurred first. If tumor progression data included more than 1 date, the first date was used.

    8. Expression Analysis of Tumor Biomarkers (HGF, EGFR, and c-Met ) at Baseline Using Immunohistochemistry (IHC) Method [Baseline]

      Tumor biomarkers such as HGF, EGFR, and c-Met were analyzed in tumor cells (neoplastic compartment) of tumor specimens from both expansion cohorts 1 and 2 by IHC. The H score was derived by summing the percentages of cells staining at each intensity multiplied by the weighted intensity of staining (0, 1+, 2+, 3+, where 3+ indicates the strongest staining, 2+ indicates medium staining, 1+ indicates weak staining, and 0 indicates no staining). Minimum calculated score of 0 to maximum calculated score of 300, where 0 correspond to no expression and maximum score of 300 indicates the strongest expression. However, the biomarker expression level (higher or lower) was not a predictor of outcome.

    9. Expression Analysis of Tumor Biomarkers (EGFR, and c-Met) at Baseline Using Fluorescent in Situ Hybridization (FISH) Method [Baseline]

      Expression of tumor biomarkers EGFR and cMet at Baseline (using FISH method) are presented in this outcome measure.

    10. Number of Participants With c-Met, HER2, EGFR Amplification and ALK Rearrangement at Baseline Using FISH Method [Baseline]

      Participants showed amplification of c-Met, HER2, and EGFR in the tumor cells and gene rearrangement of ALK are presented in this outcome measure.

    11. Plasma Concentration of sMet by Study Visits [At screening and Cycle 1 Day 1 (C1D1) (6 hours post dose), and C1D15, C2D1, C2D15 (all predose).]

      This outcome measure presented the plasma concentration of sMet at different study visits. s-Met was analyzed using an enzyme-linked immunosorbent assay (ELISA).

    12. Number of Participants With EGFR Mutation at Baseline [Baseline]

      Sample analyses were performed in accordance to Good Laboratory Practice (GLP) guidance and included mutation detection for EGFR gene.

    13. Number of Participants With KRAS Mutation (GLY12CYS) at Baseline [Baseline]

      Sample analyses were performed in accordance to GLP guidance and included mutation detection for KRAS gene.

    14. Number of Participants With PIK3CA Mutation at Baseline [Baseline]

      Sample analyses were performed in accordance to GLP guidance and included mutation detection for PIK3CA gene.

    15. Number of Participants With ROS1 Gene Translocation at Baseline [Baseline]

      Sample analyses were performed in accordance to GLP guidance and included translocation detection (RNA based) for ROS1 gene.

    16. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Area Under the Plasma Concentration-time Profile From Time Zero to the Last Quantifiable Concentration (AUClast) [Cycle 1 (C1)/Day 1 (D1), C1D15]

      At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included AUClast for crizotinib and PF-06260182. AUClast was calculated using Linear/Log trapezoidal method.

    17. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Area Under the Plasma Concentration-time Curve 10 (AUC10) [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). AUC10 was calculated using Linear/Log trapezoidal method. The below analysis table included geometric Mean and Geometric Coefficient of Variation of AUC10 for crizotinib and PF-06260182. Arithmetic mean was presented if the n=2.

    18. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Maximum Plasma Concentration (Cmax) [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Cmax for crizotinib and PF-06260182. Cmax was observed directly from data.

    19. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Time of Last Quantifiable Concentration (Tlast) [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tlast for crizotinib and PF-06260182. Tlast was observed directly from data.

    20. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase -Time to Maximum Plasma Concentration (Tmax) [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tmax for crizotinib and PF-06260182. Tmax was observed directly from data as time of first occurrence.

    21. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - AUClast [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included AUClast for dacomitinib and PF-05199265. AUClast was calculated using Linear/Log trapezoidal method.

    22. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - AUC24 [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included AUC24 for dacomitinib and PF-05199265. AUC24 was calculated using Linear/Log trapezoidal method.

    23. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - Cmax [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Cmax for dacomitinib and PF-05199265. Cmax was observed directly from data.

    24. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - Tlast [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tlast for dacomitinib and PF-05199265. Tlast was observed directly from data.

    25. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - Tmax [C1D1, C1D15]

      At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tmax for dacomitinib and PF-05199265. Tmax was observed directly from data as time of first occurrence.

    26. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - AUClast [Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)]

      Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for AUClast. AUClast was calculated using Linear/Log trapezoidal method.

    27. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - AUC10 [Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)]

      Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for AUC10. AUC10 was calculated using Linear/Log trapezoidal method.

    28. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Cmin [Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)]

      Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Cmin. Cmin was observed directly from data.

    29. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Cmax [Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)]

      Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Cmax. Cmax was observed directly from data.

    30. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Tlast [Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)]

      Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Tlast. Tlast was observed directly from data.

    31. Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Tmax [Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)]

      Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Tmax. Tmax was observed directly from data as time of first occurrence.

    32. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - AUClast [Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)]

      PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (AUClast) following dacomitinib alone and in combination with crizotinib are summarized in the below table. AUClast was calculated using Linear/Log trapezoidal method.

    33. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - AUC24 [Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)]

      PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (AUC24) following dacomitinib alone and in combination with crizotinib are summarized in the below table. AUC24 was calculated using Linear/Log trapezoidal method.

    34. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Cmin [Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)]

      PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Cmin) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Cmin was observed directly from data.

    35. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Cmax [Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)]

      PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Cmax) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Cmax was observed directly from data.

    36. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Tlast [Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)]

      PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Tlast) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Tlast was observed directly from data.

    37. Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Tmax [Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)]

      PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Tmax) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Tmax was observed directly from data as time of first occurrence.

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    18 Years and Older
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    • advanced non small cell lung cancer (dose escalation phase)

    • acquired resistance to erlotinib or gefitinib (expansion phase)

    • mandatory entrance biopsy (expansion phase)

    Exclusion Criteria:
    • interstitial lung disease

    • unstable brain metastases

    • leptomeningeal disease

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Clinical Trials Office University of Colorado Hospital (CTO) Aurora Colorado United States 80045
    2 DRUG SHIPMENT: University of Colorado Cancer Center Aurora Colorado United States 80045
    3 Rocky Mountain Lions Eye Institute Aurora Colorado United States 80045
    4 University of Colorado Hospital Aurora Colorado United States 80045
    5 CCR, National Cancer Institute Bethesda Maryland United States 20892
    6 Drug Shipment Only Boston Massachusetts United States 02114
    7 Massachusetts General Hospital Boston Massachusetts United States 02114
    8 Brigham & Women's Hospital Boston Massachusetts United States 02115
    9 Drug Shipment Only Boston Massachusetts United States 02215
    10 Peter MacCallum Cancer Centre, Division of Haematology and Medical Oncology East Melbourne Victoria Australia 3002

    Sponsors and Collaborators

    • Pfizer

    Investigators

    • Study Director: Pfizer CT.gov Call Center, Pfizer

    Study Documents (Full-Text)

    None provided.

    More Information

    Additional Information:

    Publications

    None provided.
    Responsible Party:
    Pfizer
    ClinicalTrials.gov Identifier:
    NCT01121575
    Other Study ID Numbers:
    • A8081006
    First Posted:
    May 12, 2010
    Last Update Posted:
    Oct 28, 2015
    Last Verified:
    Oct 1, 2015

    Study Results

    Participant Flow

    Recruitment Details This was a Phase 1, multicenter, open-label, non-randomized study of combined oral crizotinib and oral dacomitinib in participants with advanced non-small cell lung cancer (NSCLC). The study consisted of a dose Escalation Phase and an Expansion Phase. The Expansion Phase consisted of Expansion Cohort 1 and 2 which ran concurrently.
    Pre-assignment Detail A total of 70 participants were enrolled and received study treatment in the United States (3 centers) and Australia (1 center). 33 participants in Escalation Phase and 37 participants in Expansion Phase (25 participants in Cohort 1 and 12 participants in Cohort 2) received treatment. The last participant completed the study on 11 Feb 2014.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg twice a day (BID) and oral dacomitinib 30 mg once daily (QD). The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Period Title: Overall Study
    STARTED 14 6 7 6 25 12
    Treated 14 6 7 6 25 12
    COMPLETED 0 0 0 0 0 0
    NOT COMPLETED 14 6 7 6 25 12

    Baseline Characteristics

    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD Expansion Cohort 1 Expansion Cohort 2 Total
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone. Total of all reporting groups
    Overall Participants 14 6 7 6 25 12 70
    Age (Years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [Years]
    53.6
    (9.87)
    61.7
    (7.53)
    54.7
    (11.34)
    62.2
    (13.61)
    61.8
    (10.83)
    59.6
    (11.74)
    59.1
    (11.05)
    Sex: Female, Male (Count of Participants)
    Female
    5
    35.7%
    5
    83.3%
    6
    85.7%
    2
    33.3%
    16
    64%
    9
    75%
    43
    61.4%
    Male
    9
    64.3%
    1
    16.7%
    1
    14.3%
    4
    66.7%
    9
    36%
    3
    25%
    27
    38.6%

    Outcome Measures

    1. Primary Outcome
    Title Overview of Treatment-emergent All Causalities Adverse Events (AEs) in Escalation Phase
    Description AE was any untoward medical occurrence with study drug/ device in a trial participant. Serious adverse event (SAE) was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using National Cancer Institute (NCI) Common Terminology Criteria for AEs (CTCAE) v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.
    Time Frame Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population included all enrolled participants who had received at least 1 dose of study medication.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 14 6 7 6
    Participants with AEs
    14
    100%
    6
    100%
    7
    100%
    6
    100%
    Participants with Grade 3 or 4 AEs
    9
    64.3%
    4
    66.7%
    5
    71.4%
    5
    83.3%
    Participants with Grade 5 AEs
    3
    21.4%
    0
    0%
    0
    0%
    1
    16.7%
    Participants with SAEs
    6
    42.9%
    2
    33.3%
    1
    14.3%
    4
    66.7%
    2. Primary Outcome
    Title Overview of Treatment-emergent All Causalities AEs in Expansion Phase
    Description AE was any untoward medical occurrence with study drug/ device in a trial participant. SAE was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using NCI CTCAE v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.
    Time Frame Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population included all enrolled participants who had received at least 1 dose of study medication.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 25 12
    Participants with AEs
    22
    157.1%
    11
    183.3%
    Participants with Grade 3 or 4 AEs
    12
    85.7%
    8
    133.3%
    Participants with Grade 5 AEs
    3
    21.4%
    2
    33.3%
    Participants with SAEs
    8
    57.1%
    8
    133.3%
    3. Primary Outcome
    Title Overview of Treatment-emergent, Treatment-related AEs in Escalation Phase
    Description An AE was any untoward medical occurrence attributed to study treatment in a participant who received study treatment. SAE was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using NCI CTCAE v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.
    Time Frame Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population included all enrolled participants who had received at least 1 dose of study medication.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 14 6 7 6
    Participants with treatment-related AEs
    14
    100%
    6
    100%
    7
    100%
    6
    100%
    Participants with treatment-related Grade 3/ 4 AEs
    8
    57.1%
    3
    50%
    3
    42.9%
    3
    50%
    Participants with treatment-related Grade 5 AEs
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    Participants with treatment-related SAEs
    1
    7.1%
    2
    33.3%
    0
    0%
    1
    16.7%
    4. Primary Outcome
    Title Overview of Treatment-emergent, Treatment-related AEs in Expansion Phase
    Description An AE was any untoward medical occurrence attributed to study treatment in a participant who received study treatment. SAE was an AE resulting in death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. AEs were reported from signing of informed consent until 28 days after the last dose of study medication. SAEs were reported after this time frame if considered to be treatment related. The severity of AEs were graded by the investigator using NCI CTCAE v.4.02 and was assessed as Grade 0: no change from normal or reference range; Grade 1: mild; Grade 2: moderate; Grade 3: severe; Grade 4: life-threatening or disabling; Grade 5: death related to AE. However the below table included Grade 3, 4, and 5 AEs.
    Time Frame Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population included all enrolled participants who had received at least 1 dose of study medication.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 25 12
    Participants with treatment-related AEs
    22
    157.1%
    11
    183.3%
    Participants with treatment-related Grade 3/ 4 AEs
    7
    50%
    6
    100%
    Participants with treatment-related Grade 5 AEs
    0
    0%
    0
    0%
    Participants with treatment-related SAEs
    3
    21.4%
    3
    50%
    5. Primary Outcome
    Title Number of Participants With Dose Limiting Toxicities (DLTs) in Escalation Phase
    Description DLTs were those AEs which occurred in Cycle 1 of treatment in Dose Escalation Phase which may be attributed to study drug [combined Crizotinib (PF-02341066) plus Dacomitinib (PF-00299804)] without a clear alternative explanation and despite the use of adequate/maximal medical intervention as dictated by local institutional clinical practices or the judgment of the investigator. The following events were considered DLTs (using CTCAE version 4.02);1. Grade ≥4 hematologic events. 2. Grade ≥3 non-hematological events (except Grade 3/4 asymptomatic hypophosphatemia and Grade 3/4 hyperuricemia without signs and symptoms of gout). Nausea, vomiting or diarrhea had to have persisted at Grade 3 or 4 despite maximal medical therapy. Grade 3 hypertension will be considered a DLT only if the event is unmanageable by standard approved pharmacologic agents or if the symptomatic sequelae are identified despite appropriate medical intervention.
    Time Frame Cycle 1 (4 weeks)

    Outcome Measure Data

    Analysis Population Description
    The DLT evaluable population was defined as safety analysis (SA) participants in the Dose Escalation phase who did not have a major treatment deviation during the first cycle.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 14 6 7 6
    Grade 3 Alanine aminotransferase increased
    0
    0%
    1
    16.7%
    0
    0%
    0
    0%
    Grade 3 Diarrhoea
    0
    0%
    1
    16.7%
    0
    0%
    0
    0%
    Grade 3 Mucosal inflammation
    0
    0%
    0
    0%
    1
    14.3%
    0
    0%
    6. Secondary Outcome
    Title Number of Participants With Stable Disease and Stable Disease Duration in Escalation Phase
    Description If a participant had not achieved an objective response with confirmed complete response (CR) or partial response (PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) - 1.1 as determined by the investigators, relative to the response evaluable population, but remained stable for at least 6 weeks after first dose, then the best overall response for such a participant was considered as stable disease.
    Time Frame From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)

    Outcome Measure Data

    Analysis Population Description
    The response evaluable population included participants in safety analysis population who had an adequate Baseline tumor assessment.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 14 6 7 6
    Stable Disease
    10
    71.4%
    3
    50%
    2
    28.6%
    5
    83.3%
    0- < 3 months
    3
    21.4%
    2
    33.3%
    1
    14.3%
    0
    0%
    3- < 6 months
    6
    42.9%
    1
    16.7%
    1
    14.3%
    3
    50%
    6- < 9 months
    1
    7.1%
    0
    0%
    0
    0%
    2
    33.3%
    7. Secondary Outcome
    Title Number of Participants With Stable Disease and Stable Disease Duration in Expansion Phase
    Description If a participant had not achieved an objective response with confirmed complete response (CR) or partial response (PR) according to RECIST (1.1) as determined by the investigators, relative to the response evaluable population, but remained stable for at least 6 weeks after first dose, then the best overall response for such a participant was considered as stable disease.
    Time Frame From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)

    Outcome Measure Data

    Analysis Population Description
    The response evaluable population included participants in safety analysis population who had an adequate Baseline tumor assessment.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 22 12
    Stable Disease
    6
    42.9%
    5
    83.3%
    0- < 3 months
    1
    7.1%
    0
    0%
    3- < 6 months
    4
    28.6%
    4
    66.7%
    6- < 9 months
    1
    7.1%
    1
    16.7%
    8. Secondary Outcome
    Title Number of Participants With Objective Response Rate (ORR) in Escalation Phase
    Description ORR was defined as the percent of participants with CR or PR according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1) determined by study physicians, relative to the response evaluable population. Participants were considered non-responders until proven otherwise. Thus, participants who: 1) Did not have CR or PR while on study, or 2) Did not have a post-baseline tumor evaluation, or 3) Received anti-tumor treatment other than the study medication prior to reaching a CR or PR, or 4) Died, in progress, or drop out for any reason prior to reaching a CR or PR, were counted as non-responders in the assessment of ORR. To be assigned a status of PR or CR, changes in tumor measurements in participants with responding tumors were confirmed by repeated tumor assessment that were performed 4 weeks after the criteria for response were first met.
    Time Frame From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)

    Outcome Measure Data

    Analysis Population Description
    The response evaluable population included participants in safety analysis population who had an adequate Baseline tumor assessment.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 14 6 7 6
    Number (95% Confidence Interval) [Participants]
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    9. Secondary Outcome
    Title Number of Participants With ORR in Expansion Phase
    Description ORR was defined as the percent of participants with CR or PR according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1) determined by study physicians, relative to the response evaluable population. Participants were considered non-responders until proven otherwise. Thus, participants who: 1) Did not have CR or PR while on study, or 2) Did not have a post-baseline tumor evaluation, or 3) Received anti-tumor treatment other than the study medication prior to reaching a CR or PR, or 4) Died, in progress, or drop out for any reason prior to reaching a CR or PR, were counted as non-responders in the assessment of ORR. To be assigned a status of PR or CR, changes in tumor measurements in participants with responding tumors were confirmed by repeated tumor assessment that were performed 4 weeks after the criteria for response were first met.
    Time Frame From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)

    Outcome Measure Data

    Analysis Population Description
    The response evaluable population included participants in safety analysis population who had an adequate Baseline tumor assessment.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 22 12
    Number (95% Confidence Interval) [Participants]
    1
    7.1%
    0
    0%
    10. Secondary Outcome
    Title Duration of Response for the Only Participant Shown Partial Response in Expansion Phase
    Description This outcome measure presented the duration of response for one participant in expansion cohort 1 who showed partial response.
    Time Frame From objective response to date of progression or death due to any cause, whichever occurs first (up to post treatment follow-up as 28-35 days after last dose of study treatment)

    Outcome Measure Data

    Analysis Population Description
    This Outcome Measure was only assessed for participants with response.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 1 0
    Number [Weeks]
    6.29
    11. Secondary Outcome
    Title Progression Free Survival (PFS) in Escalation Phase
    Description PFS was defined as the time from the date of the first dose to the date of the first documentation of objective tumor progression according to RECIST 1.1 or death on study due to any cause, whichever occurred first. If tumor progression data included more than 1 date, the first date was used.
    Time Frame From randomization to objective tumor progression or death (up to post treatment follow-up as 28-35 days after last dose of study treatment)

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population included all enrolled participants who had received at least 1 dose of study medication.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 14 6 7 6
    Median (95% Confidence Interval) [Months]
    3.1
    3.0
    1.7
    4.4
    12. Secondary Outcome
    Title Progression Free Survival (PFS) in Expansion Phase
    Description PFS was defined as the time from the date of the first dose to the date of the first documentation of objective tumor progression according to RECIST 1.1 or death on study due to any cause, whichever occurred first. If tumor progression data included more than 1 date, the first date was used.
    Time Frame From randomization to objective tumor progression or death (up to post treatment follow-up as 28-35 days after last dose of study treatment)

    Outcome Measure Data

    Analysis Population Description
    The safety analysis population included all enrolled participants who had received at least 1 dose of study medication. In Expansion Cohort 1, two participants had censored reasons of "no adequate baseline", of which one participant had a censored reason "no tumor assessment data available.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 23 12
    Median (95% Confidence Interval) [Months]
    2.1
    2.1
    13. Secondary Outcome
    Title Expression Analysis of Tumor Biomarkers (HGF, EGFR, and c-Met ) at Baseline Using Immunohistochemistry (IHC) Method
    Description Tumor biomarkers such as HGF, EGFR, and c-Met were analyzed in tumor cells (neoplastic compartment) of tumor specimens from both expansion cohorts 1 and 2 by IHC. The H score was derived by summing the percentages of cells staining at each intensity multiplied by the weighted intensity of staining (0, 1+, 2+, 3+, where 3+ indicates the strongest staining, 2+ indicates medium staining, 1+ indicates weak staining, and 0 indicates no staining). Minimum calculated score of 0 to maximum calculated score of 300, where 0 correspond to no expression and maximum score of 300 indicates the strongest expression. However, the biomarker expression level (higher or lower) was not a predictor of outcome.
    Time Frame Baseline

    Outcome Measure Data

    Analysis Population Description
    The tumor analysis population included participants in safety analysis population who had a screening and on-treatment tumor biopsy for assessment of tumor biomarkers.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 23 12
    HGF (N= 19, 11)
    40.0
    67.0
    EGFR (N= 14, 8)
    193.2
    170.0
    cMet (N= 19, 11)
    125.0
    165.0
    14. Secondary Outcome
    Title Expression Analysis of Tumor Biomarkers (EGFR, and c-Met) at Baseline Using Fluorescent in Situ Hybridization (FISH) Method
    Description Expression of tumor biomarkers EGFR and cMet at Baseline (using FISH method) are presented in this outcome measure.
    Time Frame Baseline

    Outcome Measure Data

    Analysis Population Description
    The tumor analysis population included participants in safety analysis population who had a screening and on-treatment tumor biopsy for assessment of tumor biomarkers.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 23 12
    Ratio of Red to Green of EGFR (N= 11, 11)
    1.580
    1.180
    Ratio of Green to Orange for cMET (N = 19, 11)
    1.040
    1.000
    15. Secondary Outcome
    Title Number of Participants With c-Met, HER2, EGFR Amplification and ALK Rearrangement at Baseline Using FISH Method
    Description Participants showed amplification of c-Met, HER2, and EGFR in the tumor cells and gene rearrangement of ALK are presented in this outcome measure.
    Time Frame Baseline

    Outcome Measure Data

    Analysis Population Description
    The tumor analysis population included participants in safety analysis population who had a screening and on-treatment tumor biopsy for assessment of tumor biomarkers.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 23 12
    c-Met amplification (N= 19, 11)
    1
    7.1%
    0
    0%
    HER2 amplification (N= 19, 7)
    0
    0%
    0
    0%
    EGFR amplification (N= 11,11)
    2
    14.3%
    3
    50%
    ALK rearrangement (N= 19, 11)
    0
    0%
    0
    0%
    16. Secondary Outcome
    Title Plasma Concentration of sMet by Study Visits
    Description This outcome measure presented the plasma concentration of sMet at different study visits. s-Met was analyzed using an enzyme-linked immunosorbent assay (ELISA).
    Time Frame At screening and Cycle 1 Day 1 (C1D1) (6 hours post dose), and C1D15, C2D1, C2D15 (all predose).

    Outcome Measure Data

    Analysis Population Description
    The soluble protein analysis population included participants in safety analysis who had a screening or C1D1 soluble protein assessment, and at least one on-treatment soluble protein assessment (C1D14 C2D1 or C2D14).
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 21 7
    Baseline at Screening
    1353411.8
    (349746.88)
    1557000.0
    (514352.02)
    C1D1
    1519047.6
    (230995.77)
    1450500.0
    (553557.13)
    C1D15
    1483157.9
    (243904.64)
    1676666.7
    (540246.86)
    C2D1
    1525666.7
    (394836.61)
    1540000.0
    (315515.45)
    C2D15
    1564666.7
    (224367.90)
    1602500.0
    (126589.89)
    17. Secondary Outcome
    Title Number of Participants With EGFR Mutation at Baseline
    Description Sample analyses were performed in accordance to Good Laboratory Practice (GLP) guidance and included mutation detection for EGFR gene.
    Time Frame Baseline

    Outcome Measure Data

    Analysis Population Description
    The tumor analysis population included participants in safety analysis population who had a screening and on-treatment tumor biopsy for assessment of tumor biomarkers.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 23 12
    Exon 18 (G719X)
    1
    7.1%
    1
    16.7%
    Exon 19 (Deletion)
    6
    42.9%
    3
    50%
    Exon 20 (T790M)
    6
    42.9%
    3
    50%
    Exon 20 (T790M/S768I)
    1
    7.1%
    0
    0%
    Exon 20 (S768I)
    0
    0%
    1
    16.7%
    Exon 21 (L858R)
    6
    42.9%
    1
    16.7%
    18. Secondary Outcome
    Title Number of Participants With KRAS Mutation (GLY12CYS) at Baseline
    Description Sample analyses were performed in accordance to GLP guidance and included mutation detection for KRAS gene.
    Time Frame Baseline

    Outcome Measure Data

    Analysis Population Description
    The tumor analysis population included participants in safety analysis population who had a screening and on-treatment tumor biopsy for assessment of tumor biomarkers.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 23 12
    Number [Participants]
    1
    7.1%
    0
    0%
    19. Secondary Outcome
    Title Number of Participants With PIK3CA Mutation at Baseline
    Description Sample analyses were performed in accordance to GLP guidance and included mutation detection for PIK3CA gene.
    Time Frame Baseline

    Outcome Measure Data

    Analysis Population Description
    The tumor analysis population included participants in safety analysis population who had a screening and on-treatment tumor biopsy for assessment of tumor biomarkers.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 23 12
    Exon 20 (M1004I) (N= 14, 6)
    1
    7.1%
    0
    0%
    Exon 20 (Q1061K) (N= 14, 6)
    1
    7.1%
    0
    0%
    Exon 20 (H1047R) (N= 14, 6)
    2
    14.3%
    0
    0%
    Exon 9 (E545K) (N= 14, 6)
    1
    7.1%
    0
    0%
    20. Secondary Outcome
    Title Number of Participants With ROS1 Gene Translocation at Baseline
    Description Sample analyses were performed in accordance to GLP guidance and included translocation detection (RNA based) for ROS1 gene.
    Time Frame Baseline

    Outcome Measure Data

    Analysis Population Description
    The tumor analysis population included participants in safety analysis population who had a screening and on-treatment tumor biopsy for assessment of tumor biomarkers. However, number of participants analyzed in the below table included the participants evaluated for ROS1 gene translocation.
    Arm/Group Title Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    Measure Participants 6 3
    Number [Participants]
    0
    0%
    0
    0%
    21. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Area Under the Plasma Concentration-time Profile From Time Zero to the Last Quantifiable Concentration (AUClast)
    Description At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included AUClast for crizotinib and PF-06260182. AUClast was calculated using Linear/Log trapezoidal method.
    Time Frame Cycle 1 (C1)/Day 1 (D1), C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Crizotinib C1D1 - single dose (n= 11, 6, 7, 6)
    509.7
    (62)
    420.8
    (53)
    506.6
    (29)
    656.6
    (65)
    Crizotinib C1D15 multiple dose (n=8, 3, 6, 5)
    1759
    (39)
    2464
    (14)
    1732
    (35)
    1672
    (44)
    PF-06260182 C1D1 - single dose (n= 11, 6, 7, 6)
    121.3
    (76)
    103.2
    (74)
    121.6
    (29)
    220.7
    (55)
    PF-06260182 C1D15 - multiple dose (n=8, 3, 6, 5)
    382.3
    (52)
    593.5
    (53)
    440.5
    (37)
    532.0
    (68)
    22. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Area Under the Plasma Concentration-time Curve 10 (AUC10)
    Description At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). AUC10 was calculated using Linear/Log trapezoidal method. The below analysis table included geometric Mean and Geometric Coefficient of Variation of AUC10 for crizotinib and PF-06260182. Arithmetic mean was presented if the n=2.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Crizotinib C1D1 - single dose (n= 8, 3, 4, 6)
    624.9
    (57)
    500.8
    (9)
    559.6
    (26)
    655.8
    (65)
    Crizotinib C1D15 multiple dose (n= 5, 2, 6, 5)
    2000
    (43)
    2620
    (NA)
    1732
    (35)
    1644
    (44)
    PF-06260182 C1D1 - single dose (n= 8, 3, 4, 6)
    162.1
    (57)
    127.6
    (32)
    141.5
    (25)
    228.0
    (57)
    PF-06260182 C1D15 - multiple dose (n= 5, 2, 6, 5)
    452.5
    (48)
    798.0
    (NA)
    473.3
    (39)
    525.5
    (68)
    23. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Maximum Plasma Concentration (Cmax)
    Description At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Cmax for crizotinib and PF-06260182. Cmax was observed directly from data.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Crizotinib C1D1 - single dose (n= 11, 6, 7, 6)
    84.24
    (58)
    94.13
    (22)
    90.86
    (29)
    114.0
    (61)
    Crizotinib C1D15 multiple dose (n=8, 3, 6, 5)
    231.5
    (41)
    329.7
    (12)
    218.1
    (33)
    268.9
    (77)
    PF-06260182 C1D1 - single dose (n= 11, 6, 7, 6)
    18.50
    (72)
    22.37
    (26)
    20.97
    (29)
    34.33
    (61)
    PF-06260182 C1D15 - multiple dose (n=8, 3, 6, 5)
    49.73
    (50)
    78.45
    (44)
    57.56
    (34)
    83.23
    (91)
    24. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase - Time of Last Quantifiable Concentration (Tlast)
    Description At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tlast for crizotinib and PF-06260182. Tlast was observed directly from data.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Crizotinib C1D1 - single dose (n= 11, 6, 7, 6)
    9.92
    (58)
    9.33
    (22)
    9.05
    (29)
    9.75
    (61)
    Crizotinib C1D15 multiple dose (n=8, 3, 6, 5)
    9.29
    (41)
    9.42
    (25)
    9.05
    (47)
    10.2
    (35)
    PF-06260182 C1D1 - single dose (n= 11, 6, 7, 6)
    9.92
    (72)
    9.33
    (26)
    9.05
    (29)
    9.75
    (61)
    PF-06260182 C1D15 - multiple dose (n=8, 3, 6, 5)
    9.29
    (50)
    9.42
    (44)
    9.05
    (34)
    10.2
    (91)
    25. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Escalation Phase -Time to Maximum Plasma Concentration (Tmax)
    Description At each designated time point, blood samples (3 mL) for pharmacokinetic (PK) analysis of crizotinib and its metabolite, PF 06260182 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tmax for crizotinib and PF-06260182. Tmax was observed directly from data as time of first occurrence.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Crizotinib C1D1 - single dose (n= 11, 6, 7, 6)
    3.00
    (58)
    3.53
    (22)
    3.92
    (29)
    3.06
    (61)
    Crizotinib C1D15 multiple dose (n=8, 3, 6, 5)
    1.68
    (41)
    6.17
    (25)
    2.00
    (47)
    6.00
    (35)
    PF-06260182 C1D1 - single dose (n= 11, 6, 7, 6)
    4.07
    (72)
    4.00
    (26)
    3.98
    (29)
    4.99
    (61)
    PF-06260182 C1D15 - multiple dose (n=8, 3, 6, 5)
    4.03
    (50)
    6.17
    (44)
    4.09
    (34)
    6.00
    (91)
    26. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - AUClast
    Description At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included AUClast for dacomitinib and PF-05199265. AUClast was calculated using Linear/Log trapezoidal method.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Dacomitinib C1D1 - single dose (n= 10, 6, 7, 5)
    208.3
    (72)
    132.3
    (153)
    146.3
    (86)
    307.7
    (56)
    Dacomitinib C1D15 multiple dose (n= 8, 4, 6, 4)
    1339
    (47)
    2343
    (70)
    1212
    (15)
    1775
    (31)
    PF-5199265 C1D1 - single dose (n= 8, 6, 6, 6)
    27.68
    (80)
    7.438
    (115)
    10.19
    (190)
    55.00
    (75)
    PF-06260182 C1D15 - multiple dose (n= 8, 4, 6, 4)
    51.48
    (108)
    52.24
    (78)
    44.97
    (90)
    161.0
    (33)
    27. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - AUC24
    Description At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included AUC24 for dacomitinib and PF-05199265. AUC24 was calculated using Linear/Log trapezoidal method.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Dacomitinib C1D1 - single dose (n= 8, 3, 5, 5)
    252.7
    (61)
    347.9
    (26)
    223.4
    (23)
    306.6
    (57)
    Dacomitinib C1D15 multiple dose (n= 8, 4, 6, 4)
    1336
    (48)
    2334
    (69)
    1203
    (15)
    1745
    (30)
    PF-5199265 C1D1 - single dose (n= 9, 3, 5, 6)
    28.57
    (87)
    14.80
    (51)
    16.08
    (169)
    54.72
    (76)
    PF-06260182 C1D15 - multiple dose (n= 8, 4, 6, 4)
    51.37
    (108)
    51.97
    (78)
    44.72
    (90)
    158.1
    (31)
    28. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - Cmax
    Description At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Cmax for dacomitinib and PF-05199265. Cmax was observed directly from data.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Dacomitinib C1D1 - single dose (n= 10, 6, 7, 5)
    15.56
    (58)
    17.49
    (23)
    12.40
    (21)
    18.22
    (64)
    Dacomitinib C1D15 multiple dose (n= 8, 4, 6, 4)
    65.00
    (52)
    122.4
    (75)
    57.04
    (13)
    86.81
    (38)
    PF-5199265 C1D1 - single dose (n= 10, 6, 6, 7)
    1.894
    (89)
    0.9888
    (79)
    0.8833
    (130)
    3.193
    (74)
    PF-06260182 C1D15 - multiple dose (n= 8, 4, 6, 4)
    2.367
    (115)
    2.649
    (75)
    2.073
    (95)
    7.560
    (34)
    29. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - Tlast
    Description At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tlast for dacomitinib and PF-05199265. Tlast was observed directly from data.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Dacomitinib C1D1 - single dose (n= 11, 6, 7, 5)
    24.0
    (8.00)
    15.9
    (15.9)
    23.6
    (24.0)
    24.0
    Dacomitinib C1D15 multiple dose (n= 8, 4, 6, 4)
    24.00
    23.9
    23.9
    24.4
    PF-5199265 C1D1 - single dose (n= 8, 6, 6, 6)
    24.0
    15.9
    23.5
    24.1
    PF-06260182 C1D15 - multiple dose (n= 8, 4, 6, 4)
    24.0
    23.9
    23.9
    24.4
    30. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Escalation Phase - Tmax
    Description At each designated time point, blood samples (3 mL) for PK analysis of dacomitinib and its metabolite, PF 05199265 were collected in appropriately labeled collection tubes containing dipotassium ethylenediamine tetra-acetic acid (K2EDTA). The below analysis table included Tmax for dacomitinib and PF-05199265. Tmax was observed directly from data as time of first occurrence.
    Time Frame C1D1, C1D15

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib 200 mg BID/ Dacomitinib 30 mg QD Crizotinib 200 mg BID/ Dacomitinib 45 mg QD Crizotinib 250 mg BID/ Dacomitinib 30 mg QD Crizotinib 250 mg QD/ Dacomitinib 45 mg QD
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg QD. The first cycle was for 28 days thereafter, each cycle was 21 days.
    Measure Participants 11 6 7 6
    Dacomitinib C1D1 - single dose (n= 11, 6, 7, 5)
    5.99
    (8.00)
    8.03
    (15.9)
    6.00
    (24.0)
    6.17
    Dacomitinib C1D15 multiple dose (n= 8, 4, 6, 4)
    6.00
    5.09
    6.09
    5.00
    PF-5199265 C1D1 - single dose (n= 8, 6, 6, 6)
    6.95
    4.04
    6.78
    6.08
    PF-06260182 C1D15 - multiple dose (n= 8, 4, 6, 4)
    6.98
    3.95
    5.92
    2.00
    31. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - AUClast
    Description Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for AUClast. AUClast was calculated using Linear/Log trapezoidal method.
    Time Frame Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib Alone (Expansion Cohort 1) Crizotinib + Dacomitinib (Expansion Cohort 1)
    Arm/Group Description Participants received crizotinib alone continuous 200 mg BID dosing for approximately 12 days (±2 days). Participants received combined oral crizotinib (200 mg BID) and oral dacomitinib (30 mg QD) on a continuous daily schedule.
    Measure Participants 19 19
    Crizotinib (n = 16, 13)
    2223
    (53)
    1365
    (47)
    PF-06260182 (n = 16, 13)
    616.3
    (77)
    356.6
    (61)
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Crizotinib 200 mg BID/ Dacomitinib 30 mg QD, Crizotinib 200 mg BID/ Dacomitinib 45 mg QD
    Comments Statistical analysis was performed for crizotinib AUClast for participants who had both Day -1 and C2D1 data. Result for the analysis of AUClast was based on data from 11 participants. No statistical analysis was performed for PF-06260182.
    Type of Statistical Test Superiority or Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Ratio of adjust geometric mean
    Estimated Value 69.25
    Confidence Interval (2-Sided) 90%
    54.22 to 88.44
    Parameter Dispersion Type:
    Value:
    Estimation Comments Ratio of crizotinib + dacomitinib / crizotinib alone adjust geometric mean was analyzed using the mixed effect model and displayed as percentage.
    32. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - AUC10
    Description Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for AUC10. AUC10 was calculated using Linear/Log trapezoidal method.
    Time Frame Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib Alone (Expansion Cohort 1) Crizotinib + Dacomitinib (Expansion Cohort 1)
    Arm/Group Description Participants received crizotinib alone continuous 200 mg BID dosing for approximately 12 days (±2 days). Participants received combined oral crizotinib (200 mg BID) and oral dacomitinib (30 mg QD) on a continuous daily schedule. Each cycle was 21 days.
    Measure Participants 19 19
    Crizotinib (n = 11, 9)
    2167
    (56)
    1489
    (44)
    PF-06260182 (n = 11, 9)
    634.3
    (82)
    422.3
    (55)
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Crizotinib 200 mg BID/ Dacomitinib 30 mg QD, Crizotinib 200 mg BID/ Dacomitinib 45 mg QD
    Comments Statistical analysis was performed for crizotinib AUC10 for participants who had both Day -1 and C2D1 data. Result for the analysis of AUC10 was based on data from 6 participants. No statistical analysis was performed for PF-06260182.
    Type of Statistical Test Superiority or Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Ratio of adjust geometric mean
    Estimated Value 78.84
    Confidence Interval (2-Sided) 90%
    58.90 to 105.54
    Parameter Dispersion Type:
    Value:
    Estimation Comments Ratio of crizotinib + dacomitinib / crizotinib alone adjust geometric mean was analyzed using the mixed effect model and displayed as percentage.
    33. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Cmin
    Description Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Cmin. Cmin was observed directly from data.
    Time Frame Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib Alone (Expansion Cohort 1) Crizotinib + Dacomitinib (Expansion Cohort 1)
    Arm/Group Description Participants received crizotinib alone continuous 200 mg BID dosing for approximately 12 days (±2 days). Participants received combined oral crizotinib (200 mg BID) and oral dacomitinib (30 mg QD) on a continuous daily schedule. Each cycle was 21 days.
    Measure Participants 19 19
    Crizotinib (n = 16, 13)
    181.8
    (64)
    102.8
    (51)
    PF-06260182 (n = 16, 13)
    47.22
    (99)
    25.53
    (60)
    34. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Cmax
    Description Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Cmax. Cmax was observed directly from data.
    Time Frame Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib Alone (Expansion Cohort 1) Crizotinib + Dacomitinib (Expansion Cohort 1)
    Arm/Group Description Participants received crizotinib alone continuous 200 mg BID dosing for approximately 12 days (±2 days). Participants received combined oral crizotinib (200 mg BID) and oral dacomitinib (30 mg QD) on a continuous daily schedule. Each cycle was 21 days.
    Measure Participants 19 19
    Crizotinib (n = 16, 13)
    306.0
    (57)
    191.5
    (43)
    PF-06260182 (n = 16, 13)
    82.92
    (79)
    51.15
    (56)
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Crizotinib 200 mg BID/ Dacomitinib 30 mg QD, Crizotinib 200 mg BID/ Dacomitinib 45 mg QD
    Comments Statistical analysis was performed for crizotinib Cmax for participants who had both Day -1 and C2D1 data. Result for the analysis of Cmax was based on data from 11 participants. No statistical analysis was performed for PF-06260182.
    Type of Statistical Test Superiority or Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Ration of adjust geometric mean
    Estimated Value 70.60
    Confidence Interval (2-Sided) 90%
    54.71 to 91.12
    Parameter Dispersion Type:
    Value:
    Estimation Comments Ratio of crizotinib + dacomitinib / crizotinib alone adjust geometric mean was analyzed using the mixed effect model and displayed as percentage.
    35. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Tlast
    Description Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Tlast. Tlast was observed directly from data.
    Time Frame Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib Alone (Expansion Cohort 1) Crizotinib + Dacomitinib (Expansion Cohort 1)
    Arm/Group Description Participants received crizotinib alone continuous 200 mg BID dosing for approximately 12 days (±2 days). Participants received combined oral crizotinib (200 mg BID) and oral dacomitinib (30 mg QD) on a continuous daily schedule. Each cycle was 21 days.
    Measure Participants 19 19
    Crizotinib (n = 16, 13)
    9.650
    (57)
    9.000
    (43)
    PF-06260182 (n = 16, 13)
    9.650
    (79)
    9.000
    (56)
    36. Secondary Outcome
    Title Plasma Crizotinib and PF-06260182 Pharmacokinetic Parameter in Expansion Cohort 1 With or Without Co-administration of Dacomitinib - Tmax
    Description Participants were evaluated for the effect of dacomitinib on steady-state PK of crizotinib. PK of crizotinib alone was evaluated on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) after a lead in period of continuous BID dosing of crizotinib for approximately 12 days (±2 days). PK of crizotinib and dacomitinib in combination treatment was evaluated on Day 1 of Cycle 2. Blood samples for crizotinib full PK were to be collected on Day -1 and Day 1 of Cycle 2. The below table included PK data for Tmax. Tmax was observed directly from data as time of first occurrence.
    Time Frame Day -1 (crizotinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Crizotinib Alone (Expansion Cohort 1) Crizotinib + Dacomitinib (Expansion Cohort 1)
    Arm/Group Description Participants received crizotinib alone continuous 200 mg BID dosing for approximately 12 days (±2 days). Participants received combined oral crizotinib (200 mg BID) and oral dacomitinib (30 mg QD) on a continuous daily schedule. Each cycle was 21 days.
    Measure Participants 19 19
    Crizotinib (n = 16, 13)
    2.04
    (57)
    3.20
    (43)
    PF-06260182 (n = 16, 13)
    3.96
    (79)
    3.95
    (56)
    37. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - AUClast
    Description PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (AUClast) following dacomitinib alone and in combination with crizotinib are summarized in the below table. AUClast was calculated using Linear/Log trapezoidal method.
    Time Frame Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Dacomitinib Alone (Expansion Cohort 2) Crizotinib + Dacomitinib (Expansion Cohort 2)
    Arm/Group Description Participants received dacomitinib (30 mg, QD) alone in 21-day cycles until disease progression. Participants who progressed on dacomitinib were changed from single agent dacomitinib at the prevailing dose to combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles) as soon as feasible, and no later than 28 days after documentation of disease progression. The participants in this group received 30 mg QD doses of dacomitinib in combination with 200 mg BID doses of crizotinib.
    Measure Participants 10 10
    Dacomitinib (n = 6, 5)
    1016
    (45)
    1148
    (44)
    PF-05199265 (n = 7, 6)
    80.94
    (598)
    78.22
    (42)
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Crizotinib 200 mg BID/ Dacomitinib 30 mg QD, Crizotinib 200 mg BID/ Dacomitinib 45 mg QD
    Comments Statistical analysis was performed for dacomitinib AUClast for participants who had both Day -1 and C2D1 data. Result for the analysis of AUClast was based on data from 3 participants. No statistical analysis was performed for PF-05199265.
    Type of Statistical Test Superiority or Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Ratio of adjust geometric mean
    Estimated Value 117.81
    Confidence Interval (2-Sided) 90%
    64.97 to 213.61
    Parameter Dispersion Type:
    Value:
    Estimation Comments Ratio of crizotinib + dacomitinib / dacomitinib alone adjust geometric mean was analyzed using the mixed effect model and displayed as percentage.
    38. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - AUC24
    Description PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (AUC24) following dacomitinib alone and in combination with crizotinib are summarized in the below table. AUC24 was calculated using Linear/Log trapezoidal method.
    Time Frame Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Dacomitinib Alone (Expansion Cohort 2) Crizotinib + Dacomitinib (Expansion Cohort 2)
    Arm/Group Description Participants received dacomitinib (30 mg, QD) alone in 21-day cycles until disease progression. Participants who progressed on dacomitinib were changed from single agent dacomitinib at the prevailing dose to combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles) as soon as feasible, and no later than 28 days after documentation of disease progression. The participants in this group received 30 mg QD doses of dacomitinib in combination with 200 mg BID doses of crizotinib.
    Measure Participants 10 10
    Dacomitinib (n = 6, 5)
    995.7
    (45)
    1148
    (44)
    PF-05199265 (n = 7, 6)
    78.57
    (587)
    78.36
    (42)
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Crizotinib 200 mg BID/ Dacomitinib 30 mg QD, Crizotinib 200 mg BID/ Dacomitinib 45 mg QD
    Comments Statistical analysis was performed for dacomitinib AUC24 for participants who had both Day -1 and C2D1 data. Result for the analysis of AUC24 was based on data from 3 participants. No statistical analysis was performed for PF-05199265.
    Type of Statistical Test Superiority or Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Ratio of adjust geometric mean
    Estimated Value 121.90
    Confidence Interval (2-Sided) 90%
    70.20 to 211.66
    Parameter Dispersion Type:
    Value:
    Estimation Comments Ratio of crizotinib + dacomitinib / dacomitinib alone adjust geometric mean was analyzed using the mixed effect model and displayed as percentage.
    39. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Cmin
    Description PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Cmin) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Cmin was observed directly from data.
    Time Frame Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Dacomitinib Alone (Expansion Cohort 2) Crizotinib + Dacomitinib (Expansion Cohort 2)
    Arm/Group Description Participants received dacomitinib (30 mg, QD) alone in 21-day cycles until disease progression. Participants who progressed on dacomitinib were changed from single agent dacomitinib at the prevailing dose to combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles) as soon as feasible, and no later than 28 days after documentation of disease progression. The participants in this group received 30 mg QD doses of dacomitinib in combination with 200 mg BID doses of crizotinib.
    Measure Participants 10 10
    Dacomitinib (n = 6, 5)
    33.11
    (58)
    39.92
    (47)
    PF-05199265 (n = 7, 6)
    5.440
    (83)
    2.901
    (47)
    40. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Cmax
    Description PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Cmax) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Cmax was observed directly from data.
    Time Frame Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Dacomitinib Alone (Expansion Cohort 2) Crizotinib + Dacomitinib (Expansion Cohort 2)
    Arm/Group Description Participants received dacomitinib (30 mg, QD) alone in 21-day cycles until disease progression. Participants who progressed on dacomitinib were changed from single agent dacomitinib at the prevailing dose to combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles) as soon as feasible, and no later than 28 days after documentation of disease progression. The participants in this group received 30 mg QD doses of dacomitinib in combination with 200 mg BID doses of crizotinib.
    Measure Participants 10 10
    Dacomitinib (n = 6, 5)
    47.15
    (44)
    59.58
    (49)
    PF-05199265 (n = 7, 6)
    4.222
    (375)
    4.070
    (37)
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Crizotinib 200 mg BID/ Dacomitinib 30 mg QD, Crizotinib 200 mg BID/ Dacomitinib 45 mg QD
    Comments Statistical analysis was performed for dacomitinib Cmax for participants who had both Day -1 and C2D1 data. Result for the analysis of Cmax was based on data from 3 participants. No statistical analysis was performed for PF-05199265.
    Type of Statistical Test Superiority or Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Ratio of adjust geometric mean
    Estimated Value 130.57
    Confidence Interval (2-Sided) 90%
    82.46 to 206.73
    Parameter Dispersion Type:
    Value:
    Estimation Comments Ratio of crizotinib + dacomitinib / dacomitinib alone adjust geometric mean was analyzed using the mixed effect model and displayed as percentage.
    41. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Tlast
    Description PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Tlast) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Tlast was observed directly from data.
    Time Frame Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Dacomitinib Alone (Expansion Cohort 2) Crizotinib + Dacomitinib (Expansion Cohort 2)
    Arm/Group Description Participants received dacomitinib (30 mg, QD) alone in 21-day cycles until disease progression. Participants who progressed on dacomitinib were changed from single agent dacomitinib at the prevailing dose to combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles) as soon as feasible, and no later than 28 days after documentation of disease progression. The participants in this group received 30 mg QD doses of dacomitinib in combination with 200 mg BID doses of crizotinib.
    Measure Participants 10 10
    Dacomitinib (n = 6, 5)
    24.40
    (44)
    23.80
    (49)
    PF-05199265 (n = 7, 6)
    24.50
    (375)
    23.80
    (37)
    42. Secondary Outcome
    Title Plasma Dacomitinib and PF-05199265 Pharmacokinetic Parameter in Expansion Cohort 2 With or Without Co-administration of Dacomitinib - Tmax
    Description PK samples for full PK evaluation of dacomitinib were drawn on Day -1 (one day before C1D1 on which the combination treatment of crizotinib and dacomitinib started) and on C2D1. Dacomitinib and PF-05199265 PK parameter (Tmax) following dacomitinib alone and in combination with crizotinib are summarized in the below table. Tmax was observed directly from data as time of first occurrence.
    Time Frame Day -1 (dacomitinib alone), C2D1 (crizotinib + dacomitinib)

    Outcome Measure Data

    Analysis Population Description
    The PK parameter analysis population was defined as all participants in the safety analysis population who had at least 1 of the PK parameters of interest. N= Number of participants in the treatment group in the indicated population. n= Number of participants contributing to the summary statistics.
    Arm/Group Title Dacomitinib Alone (Expansion Cohort 2) Crizotinib + Dacomitinib (Expansion Cohort 2)
    Arm/Group Description Participants received dacomitinib (30 mg, QD) alone in 21-day cycles until disease progression. Participants who progressed on dacomitinib were changed from single agent dacomitinib at the prevailing dose to combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles) as soon as feasible, and no later than 28 days after documentation of disease progression. The participants in this group received 30 mg QD doses of dacomitinib in combination with 200 mg BID doses of crizotinib.
    Measure Participants 10 10
    Dacomitinib (n = 6, 5)
    16.0
    (44)
    5.92
    (49)
    PF-05199265 (n = 7, 6)
    5.90
    (375)
    4.35
    (37)

    Adverse Events

    Time Frame Up to Maximum of treatment duration + 28 days for each participant (could be 295 days)
    Adverse Event Reporting Description The same event may appear as both an AE and a SAE. However, what is presented are distinct events. The below tables included all causality SAEs (frequency threshold 0%) and AEs ( frequency threshold 5%).
    Arm/Group Title PF-02341066, 200 mg BID/ PF-00299804, 30 mg QD PF-02341066, 200 mg BID/ PF-00299804, 45 mg QD PF-02341066, 250 mg BID/ PF-00299804, 30 mg QD PF-02341066, 250 mg QD/ PF-00299804, 45 mg QD Expansion Cohort 1 Expansion Cohort 2
    Arm/Group Description Participants enrolled in dose escalation phase received combined oral crizotinib 200 mg BID and oral dacomitinib 30 mg. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants enrolled in dose escalation received combined oral crizotinib 200 mg BID and oral dacomitinib 45 mg. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants enrolled in dose escalation received combined oral crizotinib 250 mg BID and oral dacomitinib 30 mg. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants enrolled in dose escalation received combined oral crizotinib 250 mg BID and oral dacomitinib 45 mg. The first cycle was for 28 days thereafter, each cycle was 21 days. Participants enrolled in expansion cohort 1 received combined oral crizotinib and oral dacomitinib on a continuous daily schedule of crizotinib 200 mg BID + dacomitinib 30 mg QD. Each cycle was 21 days. If there was unacceptable toxicity or tolerability the dose could be adjusted to a lower combined dose. Expansion Cohort 1 was designed to examine the safety of crizotinib and dacomitinib administered concomitantly in NSCLC participants with acquired resistance to erlotinib or gefitinib. Participants were treated first with single agent dacomitinib at a dose determined by agreement between the investigator and the sponsor, and then, at progression, with combined maximum tolerated dose of crizotinib and dacomitinib (given orally on a continuous schedule in 21-day cycles of crizotinib 200 mg BID + dacomitinib 30 mg QD) as soon as feasible, and no later than 28 days after documentation of progressive disease. Expansion Cohort 2 used the same participant population as Expansion Cohort 1 and was designed to assess the clinical value of adding crizotinib to dacomitinib treatment after disease progression on dacomitinib alone.
    All Cause Mortality
    PF-02341066, 200 mg BID/ PF-00299804, 30 mg QD PF-02341066, 200 mg BID/ PF-00299804, 45 mg QD PF-02341066, 250 mg BID/ PF-00299804, 30 mg QD PF-02341066, 250 mg QD/ PF-00299804, 45 mg QD Expansion Cohort 1 Expansion Cohort 2
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total / (NaN) / (NaN) / (NaN) / (NaN) / (NaN) / (NaN)
    Serious Adverse Events
    PF-02341066, 200 mg BID/ PF-00299804, 30 mg QD PF-02341066, 200 mg BID/ PF-00299804, 45 mg QD PF-02341066, 250 mg BID/ PF-00299804, 30 mg QD PF-02341066, 250 mg QD/ PF-00299804, 45 mg QD Expansion Cohort 1 Expansion Cohort 2
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 6/14 (42.9%) 2/6 (33.3%) 1/7 (14.3%) 4/6 (66.7%) 9/25 (36%) 9/12 (75%)
    Blood and lymphatic system disorders
    Anaemia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Febrile neutropenia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Cardiac disorders
    Myocardial infarction 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Syncope 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Gastrointestinal disorders
    Diarrhoea 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 2/12 (16.7%)
    Nausea 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 1/12 (8.3%)
    General disorders
    Chest pain 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Disease progression 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 2/12 (16.7%)
    Fatigue 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Hepatobiliary disorders
    Bile duct obstruction 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Immune system disorders
    Hypersensitivity 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Infections and infestations
    Lung infection 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Pneumonia 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Sepsis 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Injury, poisoning and procedural complications
    Fall 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Hip fracture 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Investigations
    Alanine aminotransferase increased 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Blood creatinine increased 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Metabolism and nutrition disorders
    Dehydration 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 1/25 (4%) 0/12 (0%)
    Hypercalcaemia 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Hyperuricaemia 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Hyponatraemia 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Neoplasm malignant 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Tumour pain 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Respiratory, thoracic and mediastinal disorders
    Acute respiratory distress syndrome 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Acute respiratory failure 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Chronic respiratory failure 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Dyspnoea 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Hypoxia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 2/25 (8%) 0/12 (0%)
    Pleural effusion 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Pulmonary embolism 0/14 (0%) 1/6 (16.7%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Vascular disorders
    Embolism 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Hypotension 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Other (Not Including Serious) Adverse Events
    PF-02341066, 200 mg BID/ PF-00299804, 30 mg QD PF-02341066, 200 mg BID/ PF-00299804, 45 mg QD PF-02341066, 250 mg BID/ PF-00299804, 30 mg QD PF-02341066, 250 mg QD/ PF-00299804, 45 mg QD Expansion Cohort 1 Expansion Cohort 2
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 14/14 (100%) 6/6 (100%) 7/7 (100%) 6/6 (100%) 25/25 (100%) 12/12 (100%)
    Blood and lymphatic system disorders
    Anaemia 4/14 (28.6%) 1/6 (16.7%) 1/7 (14.3%) 0/6 (0%) 2/25 (8%) 1/12 (8.3%)
    Neutropenia 1/14 (7.1%) 0/6 (0%) 1/7 (14.3%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Thrombocytopenia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Cardiac disorders
    Bradycardia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 3/25 (12%) 0/12 (0%)
    Endocrine disorders
    Hypogonadism 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Eye disorders
    Dry eye 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 2/12 (16.7%)
    Erythema of eyelid 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Eye inflammation 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Eye irritation 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Eye pruritus 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Lacrimation increased 0/14 (0%) 2/6 (33.3%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Periorbital oedema 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Photopsia 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 4/25 (16%) 0/12 (0%)
    Punctate keratitis 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Pupils unequal 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Trichiasis 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Vision blurred 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Visual acuity reduced 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Visual impairment 4/14 (28.6%) 2/6 (33.3%) 3/7 (42.9%) 0/6 (0%) 5/25 (20%) 2/12 (16.7%)
    Gastrointestinal disorders
    Abdominal distension 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 3/25 (12%) 1/12 (8.3%)
    Abdominal pain 2/14 (14.3%) 1/6 (16.7%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 1/12 (8.3%)
    Abdominal pain upper 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 1/12 (8.3%)
    Ascites 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Chapped lips 4/14 (28.6%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Cheilitis 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 1/12 (8.3%)
    Constipation 3/14 (21.4%) 1/6 (16.7%) 0/7 (0%) 2/6 (33.3%) 7/25 (28%) 4/12 (33.3%)
    Diarrhoea 12/14 (85.7%) 6/6 (100%) 6/7 (85.7%) 6/6 (100%) 19/25 (76%) 10/12 (83.3%)
    Dry mouth 1/14 (7.1%) 1/6 (16.7%) 1/7 (14.3%) 0/6 (0%) 2/25 (8%) 1/12 (8.3%)
    Dyspepsia 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 9/25 (36%) 1/12 (8.3%)
    Dysphagia 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Flatulence 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Gastrointestinal pain 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Gastrooesophageal reflux disease 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Glossodynia 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Haemorrhoidal haemorrhage 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Haemorrhoids 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 1/25 (4%) 0/12 (0%)
    Lip disorder 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Lip dry 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Mouth ulceration 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Nausea 10/14 (71.4%) 4/6 (66.7%) 5/7 (71.4%) 6/6 (100%) 19/25 (76%) 10/12 (83.3%)
    Oesophageal pain 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Oral pain 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Rectal lesion 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Retching 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Stomatitis 2/14 (14.3%) 1/6 (16.7%) 1/7 (14.3%) 1/6 (16.7%) 4/25 (16%) 2/12 (16.7%)
    Vomiting 9/14 (64.3%) 2/6 (33.3%) 1/7 (14.3%) 5/6 (83.3%) 11/25 (44%) 6/12 (50%)
    General disorders
    Asthenia 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Axillary pain 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Chest discomfort 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Chest pain 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Chills 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Disease progression 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Face oedema 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Fatigue 7/14 (50%) 3/6 (50%) 3/7 (42.9%) 3/6 (50%) 13/25 (52%) 7/12 (58.3%)
    Gait disturbance 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Influenza like illness 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 2/25 (8%) 0/12 (0%)
    Local swelling 1/14 (7.1%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Mucosal inflammation 1/14 (7.1%) 1/6 (16.7%) 3/7 (42.9%) 2/6 (33.3%) 6/25 (24%) 2/12 (16.7%)
    Oedema 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 3/25 (12%) 0/12 (0%)
    Oedema peripheral 7/14 (50%) 2/6 (33.3%) 3/7 (42.9%) 1/6 (16.7%) 7/25 (28%) 4/12 (33.3%)
    Pain 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Pyrexia 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Immune system disorders
    Seasonal allergy 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Infections and infestations
    Bronchitis viral 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Candida infection 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Conjunctivitis 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Dermatitis infected 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Enterocolitis infectious 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Folliculitis 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Gastrointestinal viral infection 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Herpes zoster 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Intertrigo candida 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Omphalitis 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Oral candidiasis 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Paronychia 6/14 (42.9%) 2/6 (33.3%) 1/7 (14.3%) 3/6 (50%) 8/25 (32%) 7/12 (58.3%)
    Upper respiratory tract infection 1/14 (7.1%) 1/6 (16.7%) 1/7 (14.3%) 0/6 (0%) 2/25 (8%) 1/12 (8.3%)
    Rash pustular 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Rhinitis 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Skin infection 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 1/12 (8.3%)
    Urinary tract infection 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Viral infection 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Viral upper respiratory tract infection 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Vulvovaginal mycotic infection 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Injury, poisoning and procedural complications
    Bone contusion 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Chemical eye injury 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Fall 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Investigations
    Activated partial thromboplastin time prolonged 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Alanine aminotransferase increased 2/14 (14.3%) 2/6 (33.3%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 2/12 (16.7%)
    Aspartate aminotransferase increased 0/14 (0%) 2/6 (33.3%) 1/7 (14.3%) 1/6 (16.7%) 2/25 (8%) 3/12 (25%)
    Blood alkaline phosphatase increased 1/14 (7.1%) 1/6 (16.7%) 1/7 (14.3%) 1/6 (16.7%) 2/25 (8%) 2/12 (16.7%)
    Blood creatinine increased 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 1/6 (16.7%) 1/25 (4%) 1/12 (8.3%)
    Blood glucose increased 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Blood phosphorus decreased 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Blood testosterone decreased 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Blood uric acid increased 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Electrocardiogram QT prolonged 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Haemoglobin decreased 0/14 (0%) 0/6 (0%) 0/7 (0%) 2/6 (33.3%) 0/25 (0%) 1/12 (8.3%)
    International normalised ratio increased 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Neutrophil count decreased 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Weight decreased 3/14 (21.4%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 2/12 (16.7%)
    White blood cell count decreased 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Metabolism and nutrition disorders
    Decreased appetite 7/14 (50%) 4/6 (66.7%) 3/7 (42.9%) 3/6 (50%) 10/25 (40%) 5/12 (41.7%)
    Dehydration 1/14 (7.1%) 2/6 (33.3%) 2/7 (28.6%) 1/6 (16.7%) 3/25 (12%) 0/12 (0%)
    Hyperglycaemia 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 2/6 (33.3%) 0/25 (0%) 0/12 (0%)
    Hyperkalaemia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Hyperuricaemia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Hypoalbuminaemia 2/14 (14.3%) 1/6 (16.7%) 1/7 (14.3%) 2/6 (33.3%) 4/25 (16%) 2/12 (16.7%)
    Hypocalcaemia 2/14 (14.3%) 0/6 (0%) 1/7 (14.3%) 2/6 (33.3%) 1/25 (4%) 1/12 (8.3%)
    Hypokalaemia 1/14 (7.1%) 1/6 (16.7%) 2/7 (28.6%) 1/6 (16.7%) 3/25 (12%) 2/12 (16.7%)
    Hyponatraemia 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 2/12 (16.7%)
    Hypophosphataemia 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 3/25 (12%) 1/12 (8.3%)
    Hypomagnesaemia 2/14 (14.3%) 0/6 (0%) 2/7 (28.6%) 1/6 (16.7%) 3/25 (12%) 0/12 (0%)
    Musculoskeletal and connective tissue disorders
    Arthralgia 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Back pain 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 2/12 (16.7%)
    Flank pain 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Joint swelling 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Muscle spasms 5/14 (35.7%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 1/25 (4%) 2/12 (16.7%)
    Musculoskeletal chest pain 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 2/12 (16.7%)
    Musculoskeletal discomfort 1/14 (7.1%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Musculoskeletal pain 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Musculoskeletal stiffness 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Myalgia 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 1/12 (8.3%)
    Synovial cyst 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Pyogenic granuloma 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Tumour pain 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Nervous system disorders
    Dementia 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Dizziness 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 1/6 (16.7%) 3/25 (12%) 1/12 (8.3%)
    Dizziness postural 0/14 (0%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 0/12 (0%)
    Dysgeusia 2/14 (14.3%) 2/6 (33.3%) 0/7 (0%) 2/6 (33.3%) 3/25 (12%) 1/12 (8.3%)
    Headache 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 1/12 (8.3%)
    Hyperaesthesia 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Migraine 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Myoclonus 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Peripheral sensory neuropathy 1/14 (7.1%) 2/6 (33.3%) 0/7 (0%) 1/6 (16.7%) 2/25 (8%) 0/12 (0%)
    Psychiatric disorders
    Anxiety 1/14 (7.1%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Depression 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Insomnia 3/14 (21.4%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Personality change 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Renal and urinary disorders
    Dysuria 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Urinary hesitation 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Reproductive system and breast disorders
    Genital rash 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Vulvovaginal dryness 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Respiratory, thoracic and mediastinal disorders
    Chronic respiratory failure 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Cough 2/14 (14.3%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 3/25 (12%) 2/12 (16.7%)
    Dry throat 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Dysphonia 2/14 (14.3%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Dyspnoea 2/14 (14.3%) 0/6 (0%) 0/7 (0%) 2/6 (33.3%) 4/25 (16%) 2/12 (16.7%)
    Dyspnoea exertional 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 2/6 (33.3%) 0/25 (0%) 1/12 (8.3%)
    Epistaxis 3/14 (21.4%) 3/6 (50%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Nasal discomfort 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 1/12 (8.3%)
    Nasal disorder 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Nasal dryness 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Oropharyngeal pain 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Pleural effusion 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Pneumonitis 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 2/12 (16.7%)
    Productive cough 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Pulmonary embolism 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 3/12 (25%)
    Rhinorrhoea 1/14 (7.1%) 1/6 (16.7%) 0/7 (0%) 1/6 (16.7%) 0/25 (0%) 1/12 (8.3%)
    Upper-airway cough syndrome 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Wheezing 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Skin and subcutaneous tissue disorders
    Alopecia 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 1/12 (8.3%)
    Dermatitis acneiform 4/14 (28.6%) 1/6 (16.7%) 1/7 (14.3%) 3/6 (50%) 10/25 (40%) 2/12 (16.7%)
    Dry skin 7/14 (50%) 2/6 (33.3%) 1/7 (14.3%) 3/6 (50%) 6/25 (24%) 5/12 (41.7%)
    Erythema 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Facial wasting 0/14 (0%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Onychoclasis 2/14 (14.3%) 0/6 (0%) 0/7 (0%) 1/6 (16.7%) 1/25 (4%) 0/12 (0%)
    Pruritus 2/14 (14.3%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 4/25 (16%) 0/12 (0%)
    Rash 9/14 (64.3%) 3/6 (50%) 3/7 (42.9%) 3/6 (50%) 5/25 (20%) 7/12 (58.3%)
    Rash maculo-papular 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)
    Scab 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Skin fissures 1/14 (7.1%) 1/6 (16.7%) 1/7 (14.3%) 1/6 (16.7%) 3/25 (12%) 1/12 (8.3%)
    Skin lesion 1/14 (7.1%) 1/6 (16.7%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Skin toxicity 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Skin ulcer 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 1/12 (8.3%)
    Vascular disorders
    Deep vein thrombosis 1/14 (7.1%) 0/6 (0%) 1/7 (14.3%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Embolism 0/14 (0%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 2/25 (8%) 0/12 (0%)
    Hypotension 2/14 (14.3%) 0/6 (0%) 1/7 (14.3%) 1/6 (16.7%) 2/25 (8%) 0/12 (0%)
    Intra-abdominal haematoma 0/14 (0%) 1/6 (16.7%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Lymphoedema 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Poor venous access 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 0/25 (0%) 0/12 (0%)
    Thrombophlebitis superficial 1/14 (7.1%) 0/6 (0%) 0/7 (0%) 0/6 (0%) 1/25 (4%) 0/12 (0%)

    Limitations/Caveats

    [Not Specified]

    More Information

    Certain Agreements

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

    Pfizer has the right to review disclosures, requesting a delay of less than 60 days. Investigator will postpone single center publications until after disclosure of pooled data (all sites), less than 12 months from study completion/termination at all participating sites. Investigator may not disclose previously undisclosed confidential information other than study results.

    Results Point of Contact

    Name/Title Pfizer ClinicalTrials.gov Call Center
    Organization Pfizer, Inc.
    Phone 1-800-718-1021
    Email ClinicalTrials.gov_Inquiries@pfizer.com
    Responsible Party:
    Pfizer
    ClinicalTrials.gov Identifier:
    NCT01121575
    Other Study ID Numbers:
    • A8081006
    First Posted:
    May 12, 2010
    Last Update Posted:
    Oct 28, 2015
    Last Verified:
    Oct 1, 2015