Study Of Palbociclib Combined With Chemotherapy In Pediatric Patients With Recurrent/Refractory Solid Tumors

Sponsor
Pfizer (Industry)
Overall Status
Recruiting
CT.gov ID
NCT03709680
Collaborator
Children's Oncology Group (COG) (Other)
184
84
3
69.7
2.2
0

Study Details

Study Description

Brief Summary

A study to learn about safety and find out maximum tolerable dose of palbociclib given in combination with chemotherapy (temozolomide with irinotecan or topotecan with cyclophosphamide) in children, adolescents and young adults with recurrent or refractory solid tumors (phase 1). Phase 2 to learn about the efficacy of palbociclib in combination with irinotecan and temozolomide when compared with irinotecan and temozolomide alone in the treatment of children, adolescents, and young adults with recurrent or refractory Ewing sarcoma (EWS).

Study Design

Study Type:
Interventional
Anticipated Enrollment :
184 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Intervention Model Description:
Phase 1 portion: The palbociclib plus irinotecan and temozolomide combination part of the study will comprise of a dose escalation cohort (following a rolling 6 design), a dose expansion cohort, and if applicable, other solid tumor-specific cohorts. The palbociclib plus topotecan and cyclophosphamide will comprise of a dose determination cohort(following a modified rolling 6 design), a dose expansion cohort, and if applicable, other solid tumor-specific cohorts. Phase 2 open-label, randomized portion of the study will randomize patients in a 2:1 ratio to receive either palbociclib in combination with irinotecan and temozolomide (Arm A) or irinotecan and temozolomide chemotherapy alone (Arm B). Randomization will be stratified using block randomization by type and time of current disease recurrence (primary refractory or 1st recurrence < 2 years versus 1st recurrence ≥ 2 years or 2nd or greater recurrence).Phase 1 portion: The palbociclib plus irinotecan and temozolomide combination part of the study will comprise of a dose escalation cohort (following a rolling 6 design), a dose expansion cohort, and if applicable, other solid tumor-specific cohorts. The palbociclib plus topotecan and cyclophosphamide will comprise of a dose determination cohort(following a modified rolling 6 design), a dose expansion cohort, and if applicable, other solid tumor-specific cohorts. Phase 2 open-label, randomized portion of the study will randomize patients in a 2:1 ratio to receive either palbociclib in combination with irinotecan and temozolomide (Arm A) or irinotecan and temozolomide chemotherapy alone (Arm B). Randomization will be stratified using block randomization by type and time of current disease recurrence (primary refractory or 1st recurrence < 2 years versus 1st recurrence ≥ 2 years or 2nd or greater recurrence).
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
PHASE 1/2 STUDY TO EVALUATE PALBOCICLIB (IBRANCE®) IN COMBINATION WITH IRINOTECAN AND TEMOZOLOMIDE OR IN COMBINATION WITH TOPOTECAN AND CYCLOPHOSPHAMIDE IN PEDIATRIC PATIENTS WITH RECURRENT OR REFRACTORY SOLID TUMORS
Actual Study Start Date :
May 24, 2019
Anticipated Primary Completion Date :
Jul 28, 2024
Anticipated Study Completion Date :
Mar 15, 2025

Arms and Interventions

Arm Intervention/Treatment
Experimental: Phase 2 Arm A

Palbociclib in combination with irinotecan and temozolomide.

Drug: Palbociclib
Phase 1: Administered (oral) at 55 mg/m2, 75 mg/m2, or 40 mg/m2, or 95 mg/m2 or 115 mg/m2 on days 1-14 of a 21-day cycle Phase 2 : Administered (oral) at 75 mg/m2 on days 1-14 of a 21-day cycle
Other Names:
  • Ibrance
  • Drug: Temozolomide
    Phase 1 and Phase 2: Administered at 100 mg/m2 (oral or intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Temodar
  • Drug: Irinotecan
    Phase 1 and Phase 2: Administered at 50 mg/m2 (intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Campto
  • Experimental: Phase 1

    Palbociclib in combination with temozolomide and irinotecan and/or with topotecan and cyclophosphamide.

    Drug: Palbociclib
    Phase 1: Administered (oral) at 55 mg/m2, 75 mg/m2, or 40 mg/m2, or 95 mg/m2 or 115 mg/m2 on days 1-14 of a 21-day cycle Phase 2 : Administered (oral) at 75 mg/m2 on days 1-14 of a 21-day cycle
    Other Names:
  • Ibrance
  • Drug: Temozolomide
    Phase 1 and Phase 2: Administered at 100 mg/m2 (oral or intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Temodar
  • Drug: Irinotecan
    Phase 1 and Phase 2: Administered at 50 mg/m2 (intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Campto
  • Drug: Topotecan
    Phase 1 only : Administered at 0.75 mg/m2 (intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Hycamtin
  • Drug: Cyclophosphamide
    Phase 1 only: Administered at 250 mg/m2 (intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Cytoxan
  • Active Comparator: Phase 2 Arm B

    Irinotecan and temozolomide alone.

    Drug: Temozolomide
    Phase 1 and Phase 2: Administered at 100 mg/m2 (oral or intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Temodar
  • Drug: Irinotecan
    Phase 1 and Phase 2: Administered at 50 mg/m2 (intravenous), on days 1-5 of a 21-day cycle
    Other Names:
  • Campto
  • Outcome Measures

    Primary Outcome Measures

    1. Phase 2 open-label, randomized: Event-free survival (EFS) based on Investigator assessment. [Baseline to Month 24.]

      EFS is defined as the time from randomization until first event (ie, progression, recurrence following response, second malignancy or death without progression or recurrence).

    2. Phase 1: First Cycle Dose-Limiting Toxicities (DLT) [First cycle (cycle length is approximately 21 days)]

      For Dose Escalation/Determination Part: DLT defined as any of the following events occurring during the first treatment cycle and considered at least possibly-related to study medication:Grade 4 neutropenia lasting greater than 7 days;Grade 4 thrombocytopenia lasting greater than 7 days or need for platelet transfusion for a platelet count of < 20,000 per cubic millimeters twice within a 7-day period;greater than 14-day delay in the start of a subsequent course because of neutropenia or thrombocytopenia;Grade 3 or greater non-hematologic toxicities despite optimal treatment;any Grade 2 or greater non-hematologic toxicity requiring discontinuation or interruption of palbociclib for 7 or more consecutive days during the first cycle or any grade non-hematologic toxicity that delays the start of Cycle 2 by more than 14 days;clinically significant non-hematologic laboratory test abnormality Grade 3 or greater not resolving to Grade 1 or baseline within 7 days.

    3. Phase 1: Dose Expansion Parts: Frequency of adverse events [At least 28 days after last dose]

      For Dose Expansion and Tumor-Specific Expansion Parts: Adverse events to be reported during treatment and for at least 28 days after last dose.

    4. Phase 1: Dose Expansion Parts: Percentage of Participants With Complete Response or Partial Response [Through the end of treatment (up to at least 28 days after last dose)]

      For Dose Expansion and Tumor-Specific Expansion Parts: patients with confirmed Complete Response or Partial Response per Response Evaluation Criteria in Solid Tumors (RECIST, v.1.1) or modified Response Assessment in Neuro-Oncology (RANO) for central nervous system malignancies, or International Neuroblastoma Response Criteria (INRC) for neuroblastoma, during study treatment, assessed approximately every 2 to 4 cycles (each cycle is approximately 21 days).

    Secondary Outcome Measures

    1. Phase 1 and Phase 2: Frequency of adverse events [At least 28 days after last dose]

      Adverse events to be reported during treatment and for at least 28 days after last dose.

    2. Phase 1 and Phase 2: Percentage of participants with laboratory abnormalities [At least 28 days after last dose]

      Magnesium, Calcium, Creatinine, Albumin, Alanine aminotransferase, Aspartate aminotransferase, Glucose, Phosphorus, Total Bilirubin, Blood urea nitrogen, Alkaline phosphatase, Sodium, Potassium, Chloride, Platelet Count, White Blood Cell Count (differential), hemoglobin, INR or prothrombin Time, HbA1c

    3. Phase 1 and Phase 2: Number of Participants With Clinically Significant Treatment-emergent Electrocardiogram (ECG) Findings [At least 28 days after last dose]

      Clinically significant ECG findings included: corrected QT (QTc) > 450 ms, QTc >500 ms, change in QTc between 30 and 60 ms, change in QTc greater than or equal to 60 ms.

    4. Phase 1 and Phase 2: Number of Participants With Clinically Significant Change From Baseline in Vital Signs [At least 28 days after last dose]

      systolic and diastolic blood pressure, pulse

    5. Phase 2 open-label, randomized: Event-free survival (EFS) assessed by an independent review committee. [Baseline to Month 24.]

      EFS is defined as the time from randomization until first event (ie, progression, recurrence following response, second malignancy or death without progression or recurrence).

    6. Phase 1 and Phase 2: Percentage of Participants With Complete Response or Partial Response [Through the end of treatment (up to at least 28 days after last dose)]

      Patients with confirmed Complete Response or Partial Response per Response Evaluation Criteria in Solid Tumors (RECIST, v.1.1) or modified Response Assessment in Neuro-Oncology (RANO) for central nervous system malignancies, or International Neuroblastoma Response Criteria (INRC) for neuroblastoma, during study treatment, assessed approximately every 2 to 4 cycles (each cycle is approximately 21 days).

    7. Phase 1 and Phase 2: Duration of response (DoR) for Participants Who Achieved Complete Response or Partial Response [Up to 2 years]

      DoR defined as time from date of first response (Complete Response or Partial Response) in responders to date of progression or death

    8. Phase 1 and Phase 2: Progression Free Survival (PFS) [Up to 2 years]

      PFS defined as time from date of enrollment to earliest date of the death or progressive disease

    9. Phase 1 and Phase 2: Overall Survival (OS) [Up to 2 years]

      OS defined as the time from enrollment to date of death due to any cause.

    10. Phase 2 : comparison of PET-CT response assessment to objective response on CT/MRI. [up to completion of Cycle 4 ( 12 weeks of therapy)]

      PET-CT response assessment will be compared to objective response on MRI/CT, as data permit.

    11. Phase 2: the impact of the combination of palbociclib with TMZ and IRN treatment on the quality of life (QoL) of patients with refractory or recurrent EWS. [Up to Cycle 5 (completion of 12 weeks of treatment)]

      Results of QoL reported by patient at baseline and after 2 and 4 cycles using age-appropriate PROMIS tools will be summarized for both treatment arms, as data permit. Days of hospitalization will be compared in both treatment arms.

    12. Phase 1 and Phase 2:Palbociclib Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) [PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    13. Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Maximum plasma concentration time (Tmax) [PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    14. Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) [PK sampling at time points during Cycle 1 (day 2,5, 6,14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    15. Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) [PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    16. Phase 1 and Phase 2: Palbociclib Pharmacokinetics, Oral plasma clearance (CL/F) [PK sampling at time points during Cycle 1 (day 2, 5, 6, 14) and Cycle 2 (day 5 and 14). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    17. Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) [PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    18. Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Maximum plasma concentration time (Tmax) [PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    19. Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) [PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    20. Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) [PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    21. Phase 1 and Phase 2: Temozolomide Pharmacokinetics, Oral plasma clearance (CL/F) [PK sampling at time points during Cycle 1 (day 5) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    22. Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    23. Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Maximum plasma concentration time (Tmax) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    24. Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    25. Phase 1 and Phase 2: Irinotecan (and active metabolites), Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    26. Phase 1 and Phase 2: Irinotecan (and active metabolites) Pharmacokinetics, Oral plasma clearance (CL/F) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    27. Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Oral plasma clearance (CL/F) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    28. Phase 1: Topotecan (and active metabolites), Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    29. Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    30. Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Maximum plasma concentration time (Tmax) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    31. Phase 1: Topotecan (and active metabolites) Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    32. Phase 1: Cyclophosphamide Pharmacokinetics, Oral plasma clearance (CL/F) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    33. Phase 1: Cyclophosphamide Pharmacokinetics, Maximum observed plasma concentration during a dosing interval at steady state (Css, max) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    34. Phase 1: Cyclophosphamide Pharmacokinetics, Maximum plasma concentration time (Tmax) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    35. Phase 1: Cyclophosphamide Pharmacokinetics, Area under the concentration-time curve during a dosing interval at steady state (AUCss,t) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    36. Phase 1: Cyclophosphamide Pharmacokinetics, Measured concentration at the end of a dosing interval at steady state (taken directly before next administration) (Css,trough) [PK sampling at time points during Cycle 1 (day 2, 5 and 6) and Cycle 2 (day 5). Each cycle is 21 days.]

      Multiple Dose (assuming steady state is achieved), as data permit

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    2 Years to 20 Years
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion:
    1. Histologically confirmed relapsed or refractory solid tumor as follows:
    • For dose escalation and dose determination parts: Histologically confirmed relapsed or refractory solid tumor (including CNS tumors but not lymphomas). Patients with Diffuse Intrinsic Pontine Glioma do not require histological only radiographic confirmed relapse to enroll.

    • For dose expansion and tumor specific cohorts: Histologically confirmed relapsed or refractory solid tumor including but not limited to EWS, rhabdoid tumor, rhabdomyosarcoma, neuroblastoma, and medulloblastoma. Patients with Diffuse Intrinsic Pontine Glioma do not require histological only radiographic confirmed relapse to enroll. EWS is not eligible for TOPO and CTX tumor-specific cohorts.

    • For randomized Phase 2 part: Histologically confirmed Ewing sarcoma at diagnosis or at relapse, with presence of EWSR1-ETS or FUS-ETS rearrangement. Histopathology confirmation of both EWSR1-ETS or FUS-ETS rearrangement partners is required OR availability of formalin fixed paraffin embedded (FFPE) tumor tissue sample for central testing. Patient must have relapsed or have refractory disease and at least evaluable disease in at least one site other than bone marrow that can be followed by imaging.

    1. Age ≥2 and <21 years at the time of study entry.

    2. Lansky performance status ≥50% for patients ≤16 years of age, or Eastern Cooperative Oncology Group (ECOG) 0, 1 or 2 for patients >16 years of age.

    3. Adequate bone marrow function.

    • Absolute neutrophil count ≥1000/mm3;

    • Platelet count ≥100,000/mm3 (transfusion independent, no platelet transfusion in past 7 days prior study entry);

    • Hemoglobin ≥8.5 g/dL (transfusion allowed).

    1. Adequate renal function: Serum creatinine level based on age/gender must within protocol specified limits.

    2. Adequate liver function, including:

    • Aspartate aminotransferase (AST) and alanine aminotransferase (ALT)

    ≤2.5 × upper limit of normal (ULN) or ≤5 × ULN for age, if attributable to disease involvement of the liver;

    • Total bilirubin ≤1.5 × ULN for age, unless the patient has documented Gilbert's syndrome.
    1. Patients enrolled to Phase 1 portion of the study and tumor specific cohorts must have measurable disease as defined by RECIST version 1.1 or modified RANO criteria for CNS disease or INRC for neuroblastoma. Patients with EWS enrolled to Phase 2 portion of the study are eligible with evaluable disease (eg, bone only disease with no soft tissue component).

    2. Recovered to CTCAE Grade ≤1, or to baseline, from any non-hematological acute toxicities of prior surgery, chemotherapy, immunotherapy, radiotherapy, differentiation therapy or biologic therapy, with the exception of alopecia.

    3. Serum/urine pregnancy test (for all girls ≥8 years of age) negative at screening and at the baseline visit.

    Exclusion:
    1. Phase 1 and tumor specific cohorts: For palbociclib with IRN and TMZ combination, prior treatment with a CDK4/6 inhibitor or progression while on treatment with an IRN-containing regimen that includes TMZ. Patients who have received the combination of IRN and TMZ and did not progress while on these medications are eligible. For patients enrolling in the palbociclib with TOPO and CTX combination, prior treatment with a CDK4/6 inhibitor or progression while on treatment with a TOPO-containing regimen that includes CTX. Patients who have received the combination of TOPO and CTX and did not progress while on these medications are eligible. Phase 2 portion: prior treatment with a CDK4/6 inhibitor or progression while on treatment with an IRN-containing or TMZ-containing regimen. Patients who have received IRN and/or TMZ and did not progress while on these medications are eligible.

    2. Prior intolerability to IRN and/or TMZ plus/minus palbociclib with IRN and TMZ combination and prior intolerability to TOPO and/or CTX for TOPO and CTX combination.

    3. Use of strong cytochrome P450 (CYP) 3A inhibitors or inducers. Patients who are receiving strong uridine diphosphate-glucuronosyl transferase 1A1 (UGT1A1) inhibitors within 12 days of Cycle 1 Day 1 (C1D1) are not eligible for the palbociclib with IRN and TMZ combination. Patients who are receiving strong UGT1A1 inhibitors within 12 days of C1D1 are eligible for the palbociclib with TOPO and CTX combination (See Section 5.7.1 for list of products.)

    4. Systemic anti cancer therapy within 2 weeks prior to study entry and 6 weeks for nitrosoureas.

    5. Prior irradiation to >50% of the bone marrow (see Appendix 9).

    6. Participation in other studies involving investigational drug(s) within 2 weeks or 5 half lives, whichever is longer, prior to study entry.

    7. Major surgery within 4 weeks prior to study entry. Surgical biopsies or central line placement are not considered major surgeries.

    8. For IRN and TMZ with/without palbociclib combinations: known or suspected hypersensitivity to palbociclib, IRN and/or TMZ. For combination of palbociclib with TOPO and CTX: known or suspected hypersensitivity to palbociclib, TOPO and/or CTX.

    9. Patients with known symptomatic brain tumors or brain metastases and require steroids, unless they have been on a stable or on a decreasing steroid dose for >14 days.

    10. Patients with previously diagnosed brain metastases are eligible if they have completed their prior treatment and have recovered from the acute effects of radiation therapy or surgery prior to study entry for these metastases for at least 14 days post radiation and 4 weeks post-surgery and are neurologically stable.

    11. Hereditary bone marrow failure disorder.

    12. QTc >470 msec.

    13. History of clinically significant or uncontrolled cardiac disease, including:

    • History of or active congestive heart failure; if patient had congestive heart failure resolve and >1 year from resolution, patient will be considered eligible;

    • Clinically significant ventricular arrhythmia (such as ventricular tachycardia, ventricular fibrillation or Torsades de Pointes);

    • Diagnosed or suspected congenital or acquired prolonged QT syndrome;

    • Need for medications known to prolong the QT interval;

    • Uncorrected hypomagnesemia or hypokalemia because of potential effects on the QT interval;

    • Left ventricular ejection fraction <50% or shortening fraction <28%.

    1. Recent or ongoing clinically significant gastrointestinal disorder that may interfere with absorption of orally administered drugs (eg, gastrectomy).

    2. Severe acute or chronic medical or laboratory test abnormality that may increase the risk associated with study participation or investigational product administration or may interfere with the interpretation of study results, and in the judgment of the Investigator, would make the patient inappropriate for entry into this study.

    3. Investigator site staff members directly involved in the conduct of the study and their family members, site staff members otherwise supervised by the investigator, or patients who are Pfizer employees, including their family members, directly involved in the conduct of the study.

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Children's of Alabama Birmingham Alabama United States 35233
    2 Phoenix Children's Hospital Phoenix Arizona United States 85016
    3 MemorialCare Health System - Long Beach Medical Center Long Beach California United States 90806
    4 Children's Hospital Los Angeles Los Angeles California United States 90027
    5 Children's Hospital and Research Center at Oakland Oakland California United States 94609
    6 Children's Hospital of Orange County Orange California United States 92868
    7 Lucile Packard Children's Hospital Palo Alto California United States 94304
    8 UCSF Medical Center San Francisco California United States 94158
    9 University of California San Francisco, San Francisco California United States 94158
    10 Children's Hospital Colorado Aurora Colorado United States 80045
    11 Connecticut Children's Medical Center Hartford Connecticut United States 06106
    12 Children's National Medical Center Washington District of Columbia United States 20010
    13 UF Health Shands Hospital Gainesville Florida United States 32610
    14 University of Florida College of Medicine Gainesville Florida United States 32610
    15 Arnold Palmer Hospital for Children Orlando Florida United States 32806
    16 Johns Hopkins All Children's Hospital Saint Petersburg Florida United States 33701
    17 Johns Hopkins All Children's Outpatient Care Center Saint Petersburg Florida United States 33701
    18 Johns Hopkins All Children's Hospital Tampa Florida United States 33612
    19 Children's Healthcare of Atlanta at Egleston Atlanta Georgia United States 30322
    20 Children's Healthcare of Atlanta at Scottish Rite Atlanta Georgia United States 30342
    21 Children's Healthcare of Atlanta, Medical Office Building Atlanta Georgia United States 30342
    22 Ann & Robert H. Lurie Children's Hospital of Chicago Chicago Illinois United States 60611
    23 University of Illinois College of Medicine at Peoria Peoria Illinois United States 61605
    24 Indiana University Indianapolis Indiana United States 46202-5225
    25 Norton Children's Hospital Louisville Kentucky United States 40202
    26 Novak Center for Children's Health Louisville Kentucky United States 40202
    27 Johns Hopkins University Baltimore Maryland United States 21287
    28 Dana-Farber Cancer Institute Boston Massachusetts United States 02215
    29 University of Michigan Health System Ann Arbor Michigan United States 48109
    30 University of Minnesota Masonic Children's Hospital Minneapolis Minnesota United States 55454
    31 University of Minnesota Medical Center, Fairview Minneapolis Minnesota United States 55455
    32 University of Minnesota/Masonic Cancer Center Minneapolis Minnesota United States 55455
    33 University of Minnesota Minneapolis Minnesota United States 55455
    34 University of Mississippi Medical Center Jackson Mississippi United States 39216
    35 Children's Mercy Hospital Kansas City Missouri United States 64108
    36 Washington University School of Medicine Saint Louis Missouri United States 63110
    37 Children's Hospital & Medical Center Omaha Nebraska United States 68114
    38 Hackensack University Medical Center Hackensack New Jersey United States 07601
    39 Robert Wood Johnson University Hospital New Brunswick New Jersey United States 08901
    40 Rutgers Cancer Institute of New Jersey New Brunswick New Jersey United States 08903
    41 Cohen Children's Medical Center New Hyde Park New York United States 11040
    42 Morgan Stanley Children's Hospital of New York-Presbyetrian Hospital New York New York United States 10032
    43 Cincinnati Children's Hospital Medical Center Cincinnati Ohio United States 45229
    44 University Hospitals Cleveland Medical Center Cleveland Ohio United States 44106
    45 Nationwide Children's Hospital Columbus Ohio United States 43205
    46 Cincinnati Children's Liberty Campus Liberty Township Ohio United States 45044
    47 University of Oklahoma Health Sciences Center Oklahoma City Oklahoma United States 73104
    48 Oregon Health & Science University Portland Oregon United States 97239
    49 Penn State Children's Hospital and Penn State Health Milton S. Hershey Medical Center Hershey Pennsylvania United States 17033-0850
    50 Penn State Health Milton S. Hershey Medical Center Hershey Pennsylvania United States 17033
    51 Buerger Center for Advanced Pediatric Care Philadelphia Pennsylvania United States 19104
    52 Children's Hospital of Philadelphia Philadelphia Pennsylvania United States 19104
    53 UPMC Children's Hospital of Pittsburgh Pittsburgh Pennsylvania United States 15224
    54 Children's Blood and Cancer Center Austin Texas United States 78723
    55 Dell Children's Medical Center Austin Texas United States 78723
    56 Children's Medical Center Dallas Dallas Texas United States 75235
    57 Cook Children's Medical Center Fort Worth Texas United States 76104
    58 Cook Children's H/O Infusion Center Grapevine Texas United States 76051
    59 Texas Children's Hospital Houston Texas United States 77030
    60 Children's Medical Center Plano Plano Texas United States 75024
    61 Children's Hospital of The King's Daughters Norfolk Virginia United States 23507
    62 Children's Hospital of Richmond at VCU Richmond Virginia United States 23298
    63 Seattle Children's Hospital Seattle Washington United States 98105
    64 Children's Wisconsin Milwaukee Wisconsin United States 53226
    65 Medical College of Wisconsin Milwaukee Wisconsin United States 53226
    66 Alberta Children's Hospital Calgary Alberta Canada T3B 6A8
    67 Stollery Children's Hospital Edmonton Alberta Canada T6G 2B7
    68 The Hospital for Sick Children Toronto Ontario Canada M5G 1X8
    69 CHU Sainte-Justine Montreal Quebec Canada H3T 1C5
    70 Borsod-Abaúj-Zemplén Megyei Központi Kórház és Egyetemi Oktatókórház Miskolc Borsod-abaúj-zemplén Hungary 3526
    71 Pecsi Tudomanyegyetem Klinikai Kozpont Pecs Hungary 7623
    72 All India Institute of Medical Sciences New Delhi Delhi India 110029
    73 Rajiv Gandhi Cancer Institute And Research Centre New Delhi Delhi India 110085
    74 Artemis hospital Gurugram Haryana India 122001
    75 National Cancer Center Goyang-si Kyǒnggi-do Korea, Republic of 10408
    76 Seoul National University Hospital Seoul Seoul-teukbyeolsi [seoul] Korea, Republic of 03080
    77 Asan Medical Center Seoul Seoul-teukbyeolsi [seoul] Korea, Republic of 05505
    78 Samsung Medical Center Seoul Seoul-teukbyeolsi [seoul] Korea, Republic of 06351
    79 Instytut Matki i Dziecka Warsaw Mazowieckie Poland 01-211
    80 Detska Fakultna nemocnica s poliklinikou Banska Bystrica Banska Bystrica Slovakia 974 09
    81 Narodny ustav detskych chorob Bratislava Slovakia 83340
    82 Astrid Lindgrens Barnsjukhus Stockholm Sweden 17165
    83 Ege Universitesi Hastanesi İzmir İ̇zmir Turkey 35100
    84 Hacettepe Universite Hastaneleri Ankara Turkey 06100

    Sponsors and Collaborators

    • Pfizer
    • Children's Oncology Group (COG)

    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:
    NCT03709680
    Other Study ID Numbers:
    • A5481092
    • 2021-003444-25
    First Posted:
    Oct 17, 2018
    Last Update Posted:
    Aug 24, 2022
    Last Verified:
    Aug 1, 2022

    Study Results

    No Results Posted as of Aug 24, 2022