search
Back to results

Sorafenib in Treating Young Patients With Relapsed or Refractory Solid Tumors or Leukemia

Primary Purpose

Blastic Phase, Childhood Acute Promyelocytic Leukemia With PML-RARA, Childhood Chronic Myelogenous Leukemia, BCR-ABL1 Positive

Status
Completed
Phase
Phase 1
Locations
International
Study Type
Interventional
Intervention
Laboratory Biomarker Analysis
Pharmacological Study
Sorafenib Tosylate
Sponsored by
National Cancer Institute (NCI)
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Blastic Phase

Eligibility Criteria

2 Years - 21 Years (Child, Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  • Diagnosis of 1 of the following:

    • Histologically confirmed malignant solid tumor at original diagnosis or relapse

      • Measurable or evaluable disease by CT scan or MRI
    • Histologically confirmed leukemia, including 1 of the following:

      • Acute lymphoblastic leukemia (ALL)

        • Greater than 25% blasts in the bone marrow (M3 bone marrow)
      • Acute myeloid leukemia (AML)

        • Greater than 25% blasts in the bone marrow (M3 bone marrow)
      • AML and FLT3-ITD mutation

        • Patients must have ? 5% blasts in the bone marrow
        • Active extramedullary disease (except leptomeningeal disease) allowed
      • Juvenile myelomonocytic leukemia (JMML) meeting the following criteria:

        • Peripheral blood monocytosis > 1,000/mm^3
        • Blasts (including promonocytes) are < 20% of the WBCs in the blood and of the nucleated bone marrow cells
        • No Philadelphia chromosome (Ph) or BCR/ABL fusion gene
        • Has ? 2 of the following additional diagnostic criteria:

          • Hemoglobin F increased for age
          • Immature granulocytes in the peripheral blood
          • WBC > 10,000/mm^3
          • Clonal chromosomal abnormality (e.g., may be monosomy 7)
          • Sargramostim (GM-CSF) hypersensitivity of myeloid progenitors in vitro
      • Chronic myelogenous leukemia (CML) in blast crisis

        • Greater than 25% blasts in the bone marrow (M3 bone marrow)
        • Patients with Ph-positive CML must be refractory to imatinib mesylate
  • Relapsed or refractory disease

    • Patients with acute promyelocytic leukemia (APL) must be refractory to treatment with tretinoin and arsenic trioxide
  • Standard curative therapies or therapies proven to prolong survival with an acceptable quality of life do not exist
  • Active extramedullary disease, except active leptomeningeal leukemia, allowed
  • No brain tumors or known brain metastases
  • Karnofsky performance status (PS) 50-100% (for patients > 10 years of age)
  • Lansky PS 50-100% (for patients ? 10 years of age)
  • Patients with solid tumors must have adequate bone marrow function, as defined by the following:

    • Absolute neutrophil count ? 1,000/mm^3
    • Platelet count ? 75,000/mm^3 (transfusion independent)
    • Hemoglobin ? 8.0 g/dL (red blood cell [RBC] transfusions allowed)
  • Patients with leukemia may have abnormal blood counts but must meet the following criteria:

    • Platelet count ? 20,000/mm^3 (platelet transfusions allowed)
    • Hemoglobin ? 8.0 g/L (RBC transfusions allowed)
  • Patients with acute myeloid leukemia and FLT3-ITD mutation

    • Platelet count ? 20,000/mm^3
  • Lipase and amylase normal
  • Creatinine clearance or radioisotope glomerular filtration rate ? 70 mL/min OR creatinine normal based on age as follows:

    • No greater than 0.8 mg/dL (for patients 5 years of age and under)
    • No greater than 1.0 mg/dL (for patients 6-10 years of age)
    • No greater than 1.2 mg/dL (for patients 11-15 years of age)
    • No greater than 1.5 mg/dL (for patients over 15 years of age)
  • Patients with solid tumors must meet the following criteria:

    • Bilirubin normal for age
    • ALT normal for age (for the purpose of this study, the upper limit of normal [ULN] for ALT is 45 ?/L)
    • Serum albumin ? 2 g/dL
  • Patients with leukemia must meet the following criteria:

    • Bilirubin (sum of conjugated + unconjugated) ? 1.5 times ULN for age
    • ALT ? 5.0 times ULN for age (? 225 ?/L) (for the purpose of this study, the ULN for ALT is 45 ?/L)
    • Serum albumin ? 2 g/dL
  • Albumin ? 2 g/dL
  • PT, PTT, and INR normal (for patients on prophylactic anticoagulation)
  • No evidence of dyspnea at rest
  • No exercise intolerance
  • Pulse oximetry >94% on room air, if there is clinical indication for determination
  • Diastolic blood pressure ? the 95th percentile for age and gender (height included for AML and FLT3-ITD mutation patients)
  • Not pregnant or nursing
  • Negative pregnancy test
  • Fertile patients must use effective contraception
  • No uncontrolled infection
  • Able to swallow tablets
  • No evidence of bleeding diathesis
  • No other medical condition or situation that would preclude study compliance
  • No known Gilbert syndrome
  • Fully recovered from prior chemotherapy, immunotherapy, or radiotherapy (for patients with solid tumors)
  • Recovered from the non-hematologic toxic effects of all prior therapy (for patients with leukemia)
  • Recovered from acute non-hematologic toxic effects of all prior anti-cancer chemotherapy (for patients with AML and FLT3-ITD mutation)
  • At least 7 days since prior hematopoietic growth factors
  • At least 7 days since prior biologic agents
  • At least 2 weeks since prior local palliative radiotherapy (small port)
  • At least 3 months since prior total-body irradiation, craniospinal radiotherapy, or radiation to ? 50% of the pelvis
  • At least 6 weeks since other prior substantial bone marrow radiation (e.g., skull, spine, pelvis, ribs)
  • At least 3 months since prior stem cell transplantation or rescue (for patients with solid tumors)

    • No evidence of active graft-vs-host disease
  • At least 3 months since prior myeloablative therapy followed by bone marrow or stem cell transplantation (for patients with leukemia)
  • At least 3 weeks since prior myelosuppressive chemotherapy (6 weeks for nitrosoureas) (for patients with solid tumors)

    • At least 24 hours since prior nitrosoureas (for patients with AML and FLT3-ITD mutation)
  • At least 2 weeks since prior chemotherapy (for patients with leukemia)
  • At least 3 weeks since prior monoclonal antibody therapy
  • No prior sorafenib
  • No other concurrent investigational drugs
  • No other concurrent anticancer agents or therapies, including chemotherapy, radiotherapy, immunotherapy, or biologic therapy

    • Concurrent maintenance-like chemotherapy for patients with AML and FLT3-ITD mutation allowed
  • No concurrent administration of any of the following:

    • Cytochrome P450 enzyme-inducing antiepileptic drugs (e.g., phenytoin, carbamazepine, or phenobarbital)
    • Rifampin
    • Grapefruit juice
    • Hypericum perforatum (St. John wort)
  • No concurrent therapeutic anticoagulation

    • Concurrent prophylactic anticoagulation (e.g., low-dose warfarin) of venous or arterial access devices allowed provided the requirements for PT, INR, or PTT are met

Sites / Locations

  • University of Alabama at Birmingham Cancer Center
  • Children's Hospital of Orange County
  • Lucile Packard Children's Hospital Stanford University
  • Children's National Medical Center
  • Lurie Children's Hospital-Chicago
  • Indiana University/Melvin and Bren Simon Cancer Center
  • Riley Hospital for Children
  • National Institutes of Health Clinical Center
  • Dana-Farber Cancer Institute
  • C S Mott Children's Hospital
  • University of Minnesota/Masonic Cancer Center
  • Washington University School of Medicine
  • Columbia University/Herbert Irving Cancer Center
  • State University of New York Upstate Medical University
  • Cincinnati Children's Hospital Medical Center
  • Oregon Health and Science University
  • Children's Hospital of Philadelphia
  • Children's Hospital of Pittsburgh of UPMC
  • St. Jude Children's Research Hospital
  • UT Southwestern/Simmons Cancer Center-Dallas
  • Baylor College of Medicine/Dan L Duncan Comprehensive Cancer Center
  • Seattle Children's Hospital
  • Children's Hospital of Wisconsin
  • Hospital for Sick Children
  • Centre Hospitalier Universitaire Sainte-Justine

Arms of the Study

Arm 1

Arm Type

Experimental

Arm Label

Treatment (sorafenib tosylate)

Arm Description

Patients receive oral sorafenib twice daily on days 1-28. Treatment repeats every 28 days for up to 24 courses in the absence of disease progression or unacceptable toxicity.

Outcomes

Primary Outcome Measures

Number of Patients With Treatment-related Dose Limiting Toxicity in Cycle 1 by Dose Level
Number of patients with treatment-related dose limiting toxicities in cycle 1, as defined by study protocol, stratified by dose level.
Number of Patients With Treatment-related Adverse Events
Number of patients with treatment-related adverse events stratified by dose level through study completion.
Area Under the Plasma Concentration Versus Time Curve (AUC) of Sorafenib
Area under the plasma concentration versus time curve (AUC) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Clearance (Cl) of Sorafenib
Clearance of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Half-life of Sorafenib
Half-life of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Maximum Serum Concentration (Cmax) of Sorafenib
Maximum serum concentration (Cmax) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Number of Patients With Treatment-related Dose Limiting Toxicities in Later Cycles by Dose Level
Number of patients with treatment-related dose limiting toxicities in later cycles, as defined by study protocol, stratified by dose level.
Area Under the Plasma Concentration Versus Time Curve (AUC) of Sorafenib
Area under the plasma concentration versus time curve (AUC) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.
Clearance (Cl) of Sorafenib
Clearance of Sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.
Half-life of Sorafenib
Half-life of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.
Volume of Distribution at Steady State (Vss) of Sorafenib
Volume of distribution at steady state (Vss) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors, leukemia, or AML and FLT3-ITD mutation.
Maximum Serum Concentration (Cmax) of Sorafenib
Maximum serum concentration (Cmax) of Sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.

Secondary Outcome Measures

Number of Patients Who Respond Using RECIST Criteria
Frequency (%) of patients with Partial Response (PR) or Complete Response (CR) using the RECIST criteria by study part and dose level
Mean Concentration of VEGF2
Mean concentration of VEGF2 in peripheral blood sample.
Pharmacodynamics (PD) Blood Flow Part C
Pharmacodynamics: tumor blood flow in patients with AML and FLT3-ITD mutation using dynamic contrast enhanced MRI (DEMRI) (Part C).
Number of Patients With DEMRI
Assess effect on tumor blood flow via DEMRI in patients with measurable soft tissue tumors.
Leukemia Mutations
Analyze tumor samples and leukemic blasts from patients entered on this study for the presence of ras, raf, or FLT3 (leukemia) mutations.
Plasma Inhibitory Activity (PIA)
Analyze the plasma inhibitory activity (PIA) for FLT3 phosphorylation in peripheral blood samples obtained at the time of PK studies in patients with FLT3-ITD mutation AML (Part C).

Full Information

First Posted
September 30, 2011
Last Updated
January 13, 2021
Sponsor
National Cancer Institute (NCI)
search

1. Study Identification

Unique Protocol Identification Number
NCT01445080
Brief Title
Sorafenib in Treating Young Patients With Relapsed or Refractory Solid Tumors or Leukemia
Official Title
A Phase I/II Study of the Raf Kinase and Receptor Tyrosine Kinase Inhibitor Sorafenib (BAY 43-9006, NSC# 724772) in Children With Refractory Solid Tumors or Refractory Leukemias
Study Type
Interventional

2. Study Status

Record Verification Date
January 2021
Overall Recruitment Status
Completed
Study Start Date
May 30, 2006 (Actual)
Primary Completion Date
March 16, 2012 (Actual)
Study Completion Date
December 10, 2012 (Actual)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
National Cancer Institute (NCI)

4. Oversight

Data Monitoring Committee
Yes

5. Study Description

Brief Summary
This phase I/II trial is studying the side effects and best dose of sorafenib in treating young patients with relapsed or refractory solid tumors or leukemia. Sorafenib may stop the growth of cancer cells by blocking some of the enzymes needed for cell growth and by blocking blood flow to the cancer.
Detailed Description
PRIMARY OBJECTIVES: I. Determine the maximum-tolerated dose (MTD) and recommended phase II dose of sorafenib in pediatric patients with relapsed or refractory solid tumors. II. Determine whether pediatric patients with relapsed or refractory leukemia can tolerate the MTD of sorafenib for solid tumors. III. Determine the tolerability, active N-oxide metabolite, pharmacodynamics, and activity of sorafenib a the MTD in a subset of patients with acute myeloid leukemia (AML) and FLT3-ITD mutation. IV. Determine the toxicities of this drug in these patients. V. Determine the pharmacokinetics of this drug in these patients. SECONDARY OBJECTIVES: I. Determine, preliminarily, the antitumor activity of this drug within the confines of a phase I trial. II. Assess the biologic effect of sorafenib on circulating endothelial cells (CEC), circulating CEC precursors (CECP), VEGF, and VEGF-2 in peripheral blood. III. Assess the gene expression, proteomic profile, and ERK phosphorylation in blasts of patients with refractory leukemia treated with this regimen. IV. Assess the effect of sorafenib on solid tumor vascularity and tumor blood flow using dynamic contrast-enhanced MRI (DEMRI) in patients with measurable soft tissue tumors. V. Analyze tumor samples and leukemic blasts for the presence of ras, raf, or FLT3 (leukemias) mutations. VI. Analyze the plasma inhibitory activity for FLT3 phosphorylation in peripheral blood of patients with AML and FLT3-ITD mutation. VII. Determine the tolerability, pharmacokinetics of sorafenib and sorafenib?s active N-oxide metabolite, pharmacodynamics, and activity of sorafenib administered at the MTD for refractory leukemias in a subset of patients with AML and FLT3-ITD mutation. VIII. Analyze the plasma inhibitory activity for FLT3 phosphorylation in peripheral blood samples obtained at the time of PK studies in patients with FLT3-ITD mutation AML. OUTLINE: This is a dose-escalation, multicenter study. Patients are stratified according to diagnosis (malignant solid tumor vs leukemia). STRATUM I(REFRACTORY SOLID TUMOR PATIENTS): Patients receive oral sorafenib twice daily on days 1-28. Treatment repeats every 28 days for up to 24 courses in the absence of disease progression or unacceptable toxicity. Cohorts of 3-6 patients receive escalating doses of sorafenib until the maximum-tolerated dose (MTD) is determined. The MTD is defined as the dose preceding that at which 2 of 3 or 2 of 6 patients experience dose-limiting toxicity (DLT). Once the MTD is determined, up to 6 additional patients under 12 years of age may be treated at the MTD. The MTD dose level is also expanded to enroll up to 6 patients with refractory leukemia. STRATUM II (REFRACTORY LEUKEMIA PATIENTS): A cohort of 3-6 patients with leukemia receives treatment as in stratum 1 at the MTD determined in stratum 1. If 2 of 3 or 2 of 6 patients experience a DLT at the solid tumor MTD, sorafenib is reduced by one dose level. The leukemia MTD is defined as the dose at which < 1/3 of patients experience DLT during course 1 of treatment. STRATUM III (ACUTE MYELOID LEUKEMIA AND FLT3-ITD MUTATION PATIENTS): Patients receive sorafenib as in stratum 1 at the MTD determined in stratum 2. Patients undergo blood sample collection for pharmacokinetics, pharmacodynamics (in leukemia blasts only), circulating endothelial cells (CEC), circulating CEC precursors (CECP), VEGF and VEGF-2 , gene expression, proteomic profile, ERK phosphorylation, and FLT3 phosphorylation activity. Tumor tissue samples may also be analyzed for the presence of ras, raf, or FLT3. After completion of study treatment, patients are followed periodically.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Blastic Phase, Childhood Acute Promyelocytic Leukemia With PML-RARA, Childhood Chronic Myelogenous Leukemia, BCR-ABL1 Positive, Childhood Solid Neoplasm, Chronic Myelogenous Leukemia, BCR-ABL1 Positive, Juvenile Myelomonocytic Leukemia, Philadelphia Chromosome Negative, BCR-ABL1 Positive Chronic Myelogenous Leukemia, Recurrent Childhood Acute Lymphoblastic Leukemia, Recurrent Childhood Acute Myeloid Leukemia, Recurrent Disease

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 1, Phase 2
Interventional Study Model
Single Group Assignment
Masking
None (Open Label)
Allocation
N/A
Enrollment
70 (Actual)

8. Arms, Groups, and Interventions

Arm Title
Treatment (sorafenib tosylate)
Arm Type
Experimental
Arm Description
Patients receive oral sorafenib twice daily on days 1-28. Treatment repeats every 28 days for up to 24 courses in the absence of disease progression or unacceptable toxicity.
Intervention Type
Other
Intervention Name(s)
Laboratory Biomarker Analysis
Intervention Description
Correlative studies
Intervention Type
Other
Intervention Name(s)
Pharmacological Study
Intervention Description
Correlative studies
Intervention Type
Drug
Intervention Name(s)
Sorafenib Tosylate
Other Intervention Name(s)
BAY 43-9006 Tosylate, BAY 54-9085, Nexavar, sorafenib
Intervention Description
Given orally
Primary Outcome Measure Information:
Title
Number of Patients With Treatment-related Dose Limiting Toxicity in Cycle 1 by Dose Level
Description
Number of patients with treatment-related dose limiting toxicities in cycle 1, as defined by study protocol, stratified by dose level.
Time Frame
Up to 28 days
Title
Number of Patients With Treatment-related Adverse Events
Description
Number of patients with treatment-related adverse events stratified by dose level through study completion.
Time Frame
Up to 2 years
Title
Area Under the Plasma Concentration Versus Time Curve (AUC) of Sorafenib
Description
Area under the plasma concentration versus time curve (AUC) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Time Frame
During cycle 1 or cycle 2 after at least 14 consecutive days of sorafenib administration Pre-dose, 0.5, 1, 2, 3, 5, 8 hours post dose
Title
Clearance (Cl) of Sorafenib
Description
Clearance of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Time Frame
During cycle 1 or cycle 2 after at least 14 consecutive days of sorafenib administration Pre-dose, 0.5, 1, 2, 3, 5, 8 hours post dose
Title
Half-life of Sorafenib
Description
Half-life of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Time Frame
During cycle 1 or cycle 2 after at least 14 consecutive days of sorafenib administration Pre-dose, 0.5, 1, 2, 3, 5, 8 hours post dose
Title
Maximum Serum Concentration (Cmax) of Sorafenib
Description
Maximum serum concentration (Cmax) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors or leukemias.
Time Frame
During cycle 1 or cycle 2 after at least 14 consecutive days of sorafenib administration Pre-dose, 0.5, 1, 2, 3, 5, 8 hours post dose
Title
Number of Patients With Treatment-related Dose Limiting Toxicities in Later Cycles by Dose Level
Description
Number of patients with treatment-related dose limiting toxicities in later cycles, as defined by study protocol, stratified by dose level.
Time Frame
Up to 2 years
Title
Area Under the Plasma Concentration Versus Time Curve (AUC) of Sorafenib
Description
Area under the plasma concentration versus time curve (AUC) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.
Time Frame
8 hours post dose on day 1 of cycle 1
Title
Clearance (Cl) of Sorafenib
Description
Clearance of Sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.
Time Frame
8 hours post dose on day 1 of cycle 1
Title
Half-life of Sorafenib
Description
Half-life of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.
Time Frame
8 hours post dose on day 1 of cycle 1
Title
Volume of Distribution at Steady State (Vss) of Sorafenib
Description
Volume of distribution at steady state (Vss) of sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with refractory solid tumors, leukemia, or AML and FLT3-ITD mutation.
Time Frame
8 hours post dose on day 1 of cycle 1
Title
Maximum Serum Concentration (Cmax) of Sorafenib
Description
Maximum serum concentration (Cmax) of Sorefenib administered orally as tablets, BID approximately every 12 hours for cycles of 28 days with no rest period between cycles to children with AML and FLT3-ITD mutation.
Time Frame
8 hours post dose on day 1 of cycle 1
Secondary Outcome Measure Information:
Title
Number of Patients Who Respond Using RECIST Criteria
Description
Frequency (%) of patients with Partial Response (PR) or Complete Response (CR) using the RECIST criteria by study part and dose level
Time Frame
Up to 2 years
Title
Mean Concentration of VEGF2
Description
Mean concentration of VEGF2 in peripheral blood sample.
Time Frame
28 days
Title
Pharmacodynamics (PD) Blood Flow Part C
Description
Pharmacodynamics: tumor blood flow in patients with AML and FLT3-ITD mutation using dynamic contrast enhanced MRI (DEMRI) (Part C).
Time Frame
1 week prior to enrollment, then every 28 days
Title
Number of Patients With DEMRI
Description
Assess effect on tumor blood flow via DEMRI in patients with measurable soft tissue tumors.
Time Frame
Up to 2 years
Title
Leukemia Mutations
Description
Analyze tumor samples and leukemic blasts from patients entered on this study for the presence of ras, raf, or FLT3 (leukemia) mutations.
Time Frame
1 week prior to enrollment
Title
Plasma Inhibitory Activity (PIA)
Description
Analyze the plasma inhibitory activity (PIA) for FLT3 phosphorylation in peripheral blood samples obtained at the time of PK studies in patients with FLT3-ITD mutation AML (Part C).
Time Frame
1 week prior to enrollment and then every 28 days

10. Eligibility

Sex
All
Minimum Age & Unit of Time
2 Years
Maximum Age & Unit of Time
21 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Diagnosis of 1 of the following: Histologically confirmed malignant solid tumor at original diagnosis or relapse Measurable or evaluable disease by CT scan or MRI Histologically confirmed leukemia, including 1 of the following: Acute lymphoblastic leukemia (ALL) Greater than 25% blasts in the bone marrow (M3 bone marrow) Acute myeloid leukemia (AML) Greater than 25% blasts in the bone marrow (M3 bone marrow) AML and FLT3-ITD mutation Patients must have ? 5% blasts in the bone marrow Active extramedullary disease (except leptomeningeal disease) allowed Juvenile myelomonocytic leukemia (JMML) meeting the following criteria: Peripheral blood monocytosis > 1,000/mm^3 Blasts (including promonocytes) are < 20% of the WBCs in the blood and of the nucleated bone marrow cells No Philadelphia chromosome (Ph) or BCR/ABL fusion gene Has ? 2 of the following additional diagnostic criteria: Hemoglobin F increased for age Immature granulocytes in the peripheral blood WBC > 10,000/mm^3 Clonal chromosomal abnormality (e.g., may be monosomy 7) Sargramostim (GM-CSF) hypersensitivity of myeloid progenitors in vitro Chronic myelogenous leukemia (CML) in blast crisis Greater than 25% blasts in the bone marrow (M3 bone marrow) Patients with Ph-positive CML must be refractory to imatinib mesylate Relapsed or refractory disease Patients with acute promyelocytic leukemia (APL) must be refractory to treatment with tretinoin and arsenic trioxide Standard curative therapies or therapies proven to prolong survival with an acceptable quality of life do not exist Active extramedullary disease, except active leptomeningeal leukemia, allowed No brain tumors or known brain metastases Karnofsky performance status (PS) 50-100% (for patients > 10 years of age) Lansky PS 50-100% (for patients ? 10 years of age) Patients with solid tumors must have adequate bone marrow function, as defined by the following: Absolute neutrophil count ? 1,000/mm^3 Platelet count ? 75,000/mm^3 (transfusion independent) Hemoglobin ? 8.0 g/dL (red blood cell [RBC] transfusions allowed) Patients with leukemia may have abnormal blood counts but must meet the following criteria: Platelet count ? 20,000/mm^3 (platelet transfusions allowed) Hemoglobin ? 8.0 g/L (RBC transfusions allowed) Patients with acute myeloid leukemia and FLT3-ITD mutation Platelet count ? 20,000/mm^3 Lipase and amylase normal Creatinine clearance or radioisotope glomerular filtration rate ? 70 mL/min OR creatinine normal based on age as follows: No greater than 0.8 mg/dL (for patients 5 years of age and under) No greater than 1.0 mg/dL (for patients 6-10 years of age) No greater than 1.2 mg/dL (for patients 11-15 years of age) No greater than 1.5 mg/dL (for patients over 15 years of age) Patients with solid tumors must meet the following criteria: Bilirubin normal for age ALT normal for age (for the purpose of this study, the upper limit of normal [ULN] for ALT is 45 ?/L) Serum albumin ? 2 g/dL Patients with leukemia must meet the following criteria: Bilirubin (sum of conjugated + unconjugated) ? 1.5 times ULN for age ALT ? 5.0 times ULN for age (? 225 ?/L) (for the purpose of this study, the ULN for ALT is 45 ?/L) Serum albumin ? 2 g/dL Albumin ? 2 g/dL PT, PTT, and INR normal (for patients on prophylactic anticoagulation) No evidence of dyspnea at rest No exercise intolerance Pulse oximetry >94% on room air, if there is clinical indication for determination Diastolic blood pressure ? the 95th percentile for age and gender (height included for AML and FLT3-ITD mutation patients) Not pregnant or nursing Negative pregnancy test Fertile patients must use effective contraception No uncontrolled infection Able to swallow tablets No evidence of bleeding diathesis No other medical condition or situation that would preclude study compliance No known Gilbert syndrome Fully recovered from prior chemotherapy, immunotherapy, or radiotherapy (for patients with solid tumors) Recovered from the non-hematologic toxic effects of all prior therapy (for patients with leukemia) Recovered from acute non-hematologic toxic effects of all prior anti-cancer chemotherapy (for patients with AML and FLT3-ITD mutation) At least 7 days since prior hematopoietic growth factors At least 7 days since prior biologic agents At least 2 weeks since prior local palliative radiotherapy (small port) At least 3 months since prior total-body irradiation, craniospinal radiotherapy, or radiation to ? 50% of the pelvis At least 6 weeks since other prior substantial bone marrow radiation (e.g., skull, spine, pelvis, ribs) At least 3 months since prior stem cell transplantation or rescue (for patients with solid tumors) No evidence of active graft-vs-host disease At least 3 months since prior myeloablative therapy followed by bone marrow or stem cell transplantation (for patients with leukemia) At least 3 weeks since prior myelosuppressive chemotherapy (6 weeks for nitrosoureas) (for patients with solid tumors) At least 24 hours since prior nitrosoureas (for patients with AML and FLT3-ITD mutation) At least 2 weeks since prior chemotherapy (for patients with leukemia) At least 3 weeks since prior monoclonal antibody therapy No prior sorafenib No other concurrent investigational drugs No other concurrent anticancer agents or therapies, including chemotherapy, radiotherapy, immunotherapy, or biologic therapy Concurrent maintenance-like chemotherapy for patients with AML and FLT3-ITD mutation allowed No concurrent administration of any of the following: Cytochrome P450 enzyme-inducing antiepileptic drugs (e.g., phenytoin, carbamazepine, or phenobarbital) Rifampin Grapefruit juice Hypericum perforatum (St. John wort) No concurrent therapeutic anticoagulation Concurrent prophylactic anticoagulation (e.g., low-dose warfarin) of venous or arterial access devices allowed provided the requirements for PT, INR, or PTT are met
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Brigitte C Widemann
Organizational Affiliation
COG Phase I Consortium
Official's Role
Principal Investigator
Facility Information:
Facility Name
University of Alabama at Birmingham Cancer Center
City
Birmingham
State/Province
Alabama
ZIP/Postal Code
35233
Country
United States
Facility Name
Children's Hospital of Orange County
City
Orange
State/Province
California
ZIP/Postal Code
92868
Country
United States
Facility Name
Lucile Packard Children's Hospital Stanford University
City
Palo Alto
State/Province
California
ZIP/Postal Code
94304
Country
United States
Facility Name
Children's National Medical Center
City
Washington
State/Province
District of Columbia
ZIP/Postal Code
20010
Country
United States
Facility Name
Lurie Children's Hospital-Chicago
City
Chicago
State/Province
Illinois
ZIP/Postal Code
60611
Country
United States
Facility Name
Indiana University/Melvin and Bren Simon Cancer Center
City
Indianapolis
State/Province
Indiana
ZIP/Postal Code
46202
Country
United States
Facility Name
Riley Hospital for Children
City
Indianapolis
State/Province
Indiana
ZIP/Postal Code
46202
Country
United States
Facility Name
National Institutes of Health Clinical Center
City
Bethesda
State/Province
Maryland
ZIP/Postal Code
20892
Country
United States
Facility Name
Dana-Farber Cancer Institute
City
Boston
State/Province
Massachusetts
ZIP/Postal Code
02215
Country
United States
Facility Name
C S Mott Children's Hospital
City
Ann Arbor
State/Province
Michigan
ZIP/Postal Code
48109
Country
United States
Facility Name
University of Minnesota/Masonic Cancer Center
City
Minneapolis
State/Province
Minnesota
ZIP/Postal Code
55455
Country
United States
Facility Name
Washington University School of Medicine
City
Saint Louis
State/Province
Missouri
ZIP/Postal Code
63110
Country
United States
Facility Name
Columbia University/Herbert Irving Cancer Center
City
New York
State/Province
New York
ZIP/Postal Code
10032
Country
United States
Facility Name
State University of New York Upstate Medical University
City
Syracuse
State/Province
New York
ZIP/Postal Code
13210
Country
United States
Facility Name
Cincinnati Children's Hospital Medical Center
City
Cincinnati
State/Province
Ohio
ZIP/Postal Code
45229
Country
United States
Facility Name
Oregon Health and Science University
City
Portland
State/Province
Oregon
ZIP/Postal Code
97239
Country
United States
Facility Name
Children's Hospital of Philadelphia
City
Philadelphia
State/Province
Pennsylvania
ZIP/Postal Code
19104
Country
United States
Facility Name
Children's Hospital of Pittsburgh of UPMC
City
Pittsburgh
State/Province
Pennsylvania
ZIP/Postal Code
15224
Country
United States
Facility Name
St. Jude Children's Research Hospital
City
Memphis
State/Province
Tennessee
ZIP/Postal Code
38105
Country
United States
Facility Name
UT Southwestern/Simmons Cancer Center-Dallas
City
Dallas
State/Province
Texas
ZIP/Postal Code
75390
Country
United States
Facility Name
Baylor College of Medicine/Dan L Duncan Comprehensive Cancer Center
City
Houston
State/Province
Texas
ZIP/Postal Code
77030
Country
United States
Facility Name
Seattle Children's Hospital
City
Seattle
State/Province
Washington
ZIP/Postal Code
98105
Country
United States
Facility Name
Children's Hospital of Wisconsin
City
Milwaukee
State/Province
Wisconsin
ZIP/Postal Code
53226
Country
United States
Facility Name
Hospital for Sick Children
City
Toronto
State/Province
Ontario
ZIP/Postal Code
M5G 1X8
Country
Canada
Facility Name
Centre Hospitalier Universitaire Sainte-Justine
City
Montreal
State/Province
Quebec
ZIP/Postal Code
H3T 1C5
Country
Canada

12. IPD Sharing Statement

Learn more about this trial

Sorafenib in Treating Young Patients With Relapsed or Refractory Solid Tumors or Leukemia

We'll reach out to this number within 24 hrs