Risk-Adapted Therapy for Young Children With Embryonal Brain Tumors, Choroid Plexus Carcinoma, High Grade Glioma or Ependymoma

Sponsor
St. Jude Children's Research Hospital (Other)
Overall Status
Active, not recruiting
CT.gov ID
NCT00602667
Collaborator
University of Florida (Other), National Cancer Institute (NCI) (NIH), The Pew Charitable Trusts (Other)
293
6
3
184.4
48.8
0.3

Study Details

Study Description

Brief Summary

RATIONALE: In this study a combination of anti-cancer drugs (chemotherapy) is used to treat brain tumors in young children. Using chemotherapy gives the brain more time to develop before radiation is given. The chemotherapy in this study includes the drug methotrexate. This drug was an important part of the two clinical trials which resulted in the best survival results for children less than 3 years of age with medulloblastoma. Most patients treated on this trial will also receive radiation which is carefully targeted to the area of the tumor. This type of radiation (focal conformal or proton beam radiotherapy) may result in fewer problems with thinking and learning than radiation to the whole brain and spinal cord.

PURPOSE: This clinical trial is studying how well giving combination chemotherapy together with radiation therapy works in treating young patients with newly diagnosed central nervous system tumors.

Condition or Disease Intervention/Treatment Phase
  • Drug: Induction Chemotherapy
  • Drug: Low-Risk Therapy
  • Drug: High-Risk Therapy
  • Drug: Intermediate-Risk Therapy
Phase 2

Detailed Description

All patients with medulloblastoma who were diagnosed prior to their 3rd birthday will contribute to both the biology and therapeutic primary objectives of this protocol. Furthermore patients who were ≥3 and <5 years old at the time of diagnosis will also be included in the cohort for these primary objectives as long as they meet the eligibility criteria as outlined in Amendment 8.0 of this protocol. Patients in the 3-5 year old age cohort who enrolled on previous versions of this protocol and who do not meet the criteria as outlined in Amendment 8.0 of this protocol will be excluded from the outcome analyses of the biology and therapeutic primary objectives of the protocol.

OBJECTIVES:

Primary

  • To identify patterns of methylation profiling that are associated with progression-free survival among young pediatric patients with medulloblastoma treated with risk-adapted therapy.

  • To estimate the event-free survival distribution of young medulloblastoma patients treated with risk-adapted therapy.

Secondary

  • To perform high-resolution genome-wide analyses of chromosomal abnormalities and gene expression patterns, and evaluate the relationship of these to other clinicopathological variables.

  • To evaluate specific tumor types for molecular abnormalities with suspected prognostic or therapeutic significance.

  • To evaluate the feasibility of collecting frozen and fixed tumor samples for analysis using high-resolution molecular biology tools.

  • To estimate the event-free and overall survival of patients treated with the proposed risk-adapted therapy regimen, and to descriptively compare these survival rates to historical controls.

  • To estimate the rates of local and distant disease progression in patients treated with focal radiotherapy (RT) to the post-operative tumor bed using a 5 mm clinical target volume margin.

  • To estimate the objective response rate (sustained for 8 weeks) to induction chemotherapy including high-dose intravenous methotrexate for patients with residual or metastatic disease.

  • To evaluate the feasibility and toxicity of administering low-dose intravenous vinblastine in conjunction with induction chemotherapy to patients with metastatic disease.

  • To evaluate the feasibility and toxicity of administering consolidation therapy including cyclophosphamide and pharmacokinetically targeted topotecan to patients with metastatic disease, and to estimate the sustained (for 8 weeks) objective response rate (complete response and partial response) to such therapy in patients with measurable residual disease after induction.

  • To evaluate the feasibility and toxicity of administering oral maintenance therapy in young children.

  • To use quantitative magnetic resonance (MR) measures (volumetric, diffusion, and perfusion) of young brain tumor patients receiving chemotherapy including high-dose intravenous methotrexate to assess impact of treatment on developing brain.

  • To investigate the feasibility of using PET as an in-vivo dosimetric and distal edge verification system for patients treated with proton beam therapy (for participants enrolled at St Jude only).

OUTLINE: This is a multicenter study. Patients are stratified according to disease risk (low-risk vs intermediate-risk vs high-risk). Therapy consists of risk adapted induction, consolidation and maintenance chemotherapy. Focal irradiation is given to intermediate risk patients who have reached at least 12 months of age upon completion of induction. Intermediate risk patients who have not will receive low risk chemotherapy to delay RT until the age of 12 months.

Patients may consent to provide tumor tissue and blood samples for biological studies. Tumor tissues are analyzed for the activation of the wnt signaling pathway (β-catenin), activation of the shh signaling pathway (Gli-1/SFRP1), and ERBB2; validation of novel patterns of gene expression via immunohistochemical (IHC) analysis; loss of chromosomes 6, 8p, 9q22, isochromosome 17q; amplification of MYCC, MYCN, and MYCL; validation of genetic abnormalities via interphase fluorescence in situ hybridization (iFISH); construction of gene expression profiles via microarray analysis; single nucleotide polymorphism (SNP) analysis for DNA purity and integrity using UV spectrophotometry and agarose gel electrophoresis; amplification of DNA via PCR and a combination of previously published and 'in-house' generated primers; potential oncogenes and tumor suppressor genes via DNA sequence analysis; expression of a number of cell signal proteins implicated in the biology of medulloblastoma via western blot; expression of additional proteins encoded by genes associated through SNP and gene expression array analysis with clinical disease behavior; and differential expression pattern of genes detected using microarray analysis via RT-PCR. DNA extraction and construction of tissue microarrays (TMAs) from tumor tissue will also be used for future IHC and FISH analysis. Blood samples are analyzed for constitutional DNA from patients whose tumors contain gene mutations via sequence analysis of constitutional DNA; cyclophosphamide and its metabolites via liquid chromatography mass spectroscopy method; topotecan lactone via isocratic high-performance liquid chromatography assay with fluorescence detection; and alpha-1-acid glycoprotein (AAGP) concentrations via immunoturbidimetric assay.

After completion of study treatment, patients are followed every 6 months for 5 years.

Study Design

Study Type:
Interventional
Actual Enrollment :
293 participants
Allocation:
Non-Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
Risk-Adapted Therapy for Young Children With Embryonal Brain Tumors, Choroid Plexus Carcinoma, High Grade Glioma or Ependymoma
Actual Study Start Date :
Dec 17, 2007
Actual Primary Completion Date :
Sep 27, 2017
Anticipated Study Completion Date :
Apr 30, 2023

Arms and Interventions

Arm Intervention/Treatment
Experimental: Low-Risk Patients

Patients with GTR/M0 medulloblastoma, nodular desmoplastic or high grade glioma histology will receive induction chemotherapy and low-risk therapy. Note: Accrual to the low-risk medulloblastoma cohort is closed as of 12/2/2015. Accrual to the low-risk high grade glioma remains open.

Drug: Induction Chemotherapy
All patients will receive 4 identical cycles of induction chemotherapy including highdose (5 g/m2 or 2.5g/m2 for patients less than or equal to 31 days of age at enrollment) intravenous methotrexate and standard dose vincristine, cisplatin, and cyclophosphamide.
Other Names:
  • MTX (methotrexate)
  • Oncovin(R) (vincristine)
  • Platinol-AQ(R) (cisplatin)
  • Cytoxan(R) (cyclophosphamide)
  • Drug: Low-Risk Therapy
    Induction will be followed by further conventional chemotherapy with carboplatin, cyclophosphamide, and etoposide. After consolidation, patients will receive 6 cycles of oral maintenance chemotherapy with cyclophosphamide, topotecan, and depending on the diagnosis, either erlotinib or etoposide (VP-16).
    Other Names:
  • Cytoxan(R) (cyclophosphamide)
  • Paraplatin(R) (carboplatin)
  • Vepesid(R), VP-16 (etoposide)
  • Hycamptin(R) (topotecan)
  • Tarceva(TM) (erlotinib)
  • Experimental: High-Risk Patients

    Patients with CNS metastatic disease will receive induction chemotherapy and high-risk therapy.

    Drug: Induction Chemotherapy
    All patients will receive 4 identical cycles of induction chemotherapy including highdose (5 g/m2 or 2.5g/m2 for patients less than or equal to 31 days of age at enrollment) intravenous methotrexate and standard dose vincristine, cisplatin, and cyclophosphamide.
    Other Names:
  • MTX (methotrexate)
  • Oncovin(R) (vincristine)
  • Platinol-AQ(R) (cisplatin)
  • Cytoxan(R) (cyclophosphamide)
  • Drug: High-Risk Therapy
    High risk patients will also receive vinblastine with each course of induction chemotherapy. Induction will be followed by either chemotherapy with targeted intravenous topotecan and cyclophosphamide or optional craniospinal irradiation (CSI). CSI will be offered only to patients who reach 3 years of age by the end of induction only. After consolidation, all patients will receive 6 cycles of oral maintenance chemotherapy with cyclophosphamide, topotecan, and depending on the diagnosis, either erlotinib or etoposide (VP-16).
    Other Names:
  • Velban(R) (vinblastine)
  • Cytoxan(R) (cyclophosphamide)
  • Hycamptin(R) (topotecan)
  • Tarceva(TM) (erlotinib)
  • Vepesid(R), VP-16 (etoposide)
  • Experimental: Intermediate-Risk Therapy

    Patients with M0 medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease, will receive induction chemotherapy and intermediate-risk therapy.

    Drug: Induction Chemotherapy
    All patients will receive 4 identical cycles of induction chemotherapy including highdose (5 g/m2 or 2.5g/m2 for patients less than or equal to 31 days of age at enrollment) intravenous methotrexate and standard dose vincristine, cisplatin, and cyclophosphamide.
    Other Names:
  • MTX (methotrexate)
  • Oncovin(R) (vincristine)
  • Platinol-AQ(R) (cisplatin)
  • Cytoxan(R) (cyclophosphamide)
  • Drug: Intermediate-Risk Therapy
    Induction will be followed by consolidation focal radiotherapy (RT) to the tumor bed. Patients less than 12 months old upon completion of induction will receive low risk chemotherapy to delay RT until the age of 12 months. After consolidation, patients will receive 6 cycles of oral maintenance chemotherapy with cyclophosphamide, topotecan, and depending on the diagnosis, either erlotinib or etoposide (VP-16). Note: The option to receive focal proton beam irradiation was suspended 10/29/2015. Focal photon beam irradiation continues as part of the treatment plan.
    Other Names:
  • Cytoxan(R) (cyclophosphamide)
  • Hycamptin(R) (topotecan)
  • Tarceva(TM) (erlotinib)
  • Vepesid(R), VP-16 (etoposide)
  • Outcome Measures

    Primary Outcome Measures

    1. Percent Probability of Progression-free Survival (PFS) for Medulloblastoma Patients [From date on treatment until date of first progression or relapse or disease related death or date of last contact, estimated at 1 year after treatment]

      Progression was defined as 25% increase in the size of any measurable lesion; the appearance of a new lesion; or the conversion of negative cerebrospinal fluid (CSF) cytology to positive. Defined as the time interval from date on treatment until the date of first progression, medulloblastoma-related death or date of last contact for patients who have not experienced an event. All eligible medulloblastoma patients who received any methotrexate are included in this analysis.

    2. Percent Probability of Progression-free Survival (PFS) for Medulloblastoma Patients by DNA Methylation Subgroup [From date on treatment to date of first progression or relapse or disease related death or date of last contact, estimated at 1 year after treatment]

      Defined as the time interval from date on treatment until the date of first progression, medulloblastoma-related death or date of last contact for patients who have not experienced an event. Eligible medulloblastoma patients who received any methotrexate and had molecularly confirmed medulloblastoma are included in this analysis. Five patients were excluded as 3 had no archival tissue available and 2 were found to not be medulloblastoma by methylation profile.

    3. Percent Probability of Event-free Survival (EFS) for Medulloblastoma Patients [From date on treatment to date of first progression, relapse, second malignancy or death from any cause or to date of last contact, estimated at 1 year after]

      Defined as the time interval from date on treatment until the date of first progression, second malignancy or death due to any cause; or date of last contact for patients who have not experienced an event. All eligible medulloblastoma patients who received any methotrexate are included in this analysis.

    Secondary Outcome Measures

    1. Number of Participants With Chromosomal Abnormalities [Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment]

      Amplifications and deletions (gains and losses) for chromosomes of interest are shown in the table of measured values.

    2. Numbers of Patients With Gene Alterations [Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment]

      Gene alterations, which include single nucleotide variants (SNPs), amplifications, deletions, translocations, indels, and germline alterations are shown for specific genes of interest in the results table.

    3. Numbers of Patients With Molecular Abnormalities by Tumor Type [Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment]

      Alterations included single nucleotide variants (SNPs), amplifications, deletions, translocations, indels, and germline alterations. Cytogenetic information shows gains and losses as specified in the table of measured values.

    4. Number of Successful Collections for Frozen and Fixed Tumor Samples [Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment]

      Successful collections will be defined as the number of patients who have frozen/fixed tumor samples available.

    5. Event-free Survival (EFS) Compared to Historical Controls [From date on treatment until date of first event (progression, second malignancy or death) or until date of last contact, assessed up to 10 years]

      EFS was measured from the date of initial treatment to the earliest date of disease progression, second malignancy or death for patients who fail; and to the date of last contact for patients who remain at risk for failure. 1-year EFS estimates are reported by risk group. EFS was compared to St. Jude historical cohorts by risk group using hazard ratios with 95% confidence intervals.

    6. Overall Survival (OS) Compared to Historical Controls [1 year after treatment initiation of last patient]

      OS was measured from the date of initial treatment to date of death or to date of last contact for survivors. 1-year OS estimates were reported by risk group. OS was compared to St. Jude historical cohorts by risk group using hazard ratios with 95% confidence intervals.

    7. Percentage of Patients With Objective Responses Rate to Induction Chemotherapy [From on-study date to 2 months after completion of induction chemotherapy (up to 4 months after on-study date)]

      For patients treated in the intermediate and high risk strata with residual or metastatic disease we will estimate the stratum-specific objective response rate (complete response (CR) or partial response [ PR]). All patients who receive at least 1 -dose of methotrexate are evaluable for response. Objective responses must be sustained for at least eight weeks.

    8. Feasibility and Toxicity of Administering Vinblastine With Induction Chemotherapy for Patients With Metastatic Disease as Measured by the Percentage of Courses Delayed for More Than 7 Days Due to Toxicity [From on-study date up to 4 months after on-study date]

      For the subset of patients with metastatic disease (high-risk group patients), during induction, the proportion percentage of courses during which subsequent chemotherapy administration was delayed for more than 7 days due to toxicity will be calculated. Patients were to receive 4 courses of induction and then consolidation chemotherapy.

    9. Feasibility and Toxicity of Administering Consolidation Therapy Including Cyclophosphamide and Pharmacokinetically Targeted Topotecan to Patients With Metastatic Disease Based on the Percentage of Courses Delayed for More Than 7 Days Due to Toxicity [At completion of consolidation therapy (up to 6 months after on-study date)]

      For the subset of patients with metastatic disease (high-risk group patients), during consolidation, we will calculate the number and proportion of courses during which subsequent chemotherapy administration was delayed for more than 7 days due to toxicity. Patients were to received 2 courses of consolidation chemotherapy and then maintenance therapy.

    10. Percent of Patients With Sustained Objective Responses Rate After Consolidation [8 weeks after completion of consolidation therapy (up to 8 months after on-study date)]

      For patients enrolled on the high-risk arm with measurable residual disease after induction treated with consolidation therapy, we will estimate the objective response (complete response (CR)/partial response (PR)) rate after consolidation therapy with a 95% confidence interval. Objective responses must be sustained for at least eight weeks. All patients who receive at least 1 dose of cyclophosphamide or topotecan during consolidation are evaluable for response.

    11. Feasibility and Toxicity of Administering Oral Maintenance Therapy in Children <3 Years of Age as Measured by the Percentage of Total Scheduled Maintenance Doses Received [From start of oral maintenance therapy (approximately 6 months after on-study date) to completion of oral maintenance therapy (up to 1 year after on-study date)]

      These data are based on patient diaries. For children <3 years of age, we will calculate the percentage of total scheduled doses each patient received per course for each of the oral maintenance courses and report the overall average number percentage of doses received per course across patients. If patients received all planned doses, their percentage would be 100%. If the average percentage was less than 75%, then feasibility would be in question.

    12. Change in Neurostructure, Especially White Matter Volume and Integrity [From baseline to 60 months off therapy]

      Quantitative MRI measures of change in neurostructure (especially white matter volume and integrity) over time will be assessed using a random effects model incorporating various covariates. Covariates to be considered include age at diagnosis, time since diagnosis and risk-arm. Differences in quantitative MRI measures of neurostructure volume and integrity between patient groups will be evaluated as a metric of structural neurotoxicity of therapy.

    13. Percent of PET Scans With Loss of Signal Intensity [Up to 3 times during RT consolidation]

      Measures will be analyzed for intermediate risk participants who receive proton beam therapy (PBT) and who consent. This objective aims to assess the feasibility of using post-proton beam therapy (PBT) positron emission tomography (PET) as an in-vivo dosimetric and distal edge verification system in this patient population. To quantify the decay in signal, 134 scans from 53 patients were analyzed by recording the mean activation value (MAV), the average recorded PET signal from activation, within the target volume. With each patient being given the same dose, the percent standard deviation in the MAV can serve as a quantitative representation of signal loss due to radioactive decay.

    14. Change in Concentration of Cerebrospinal Fluid (CSF) Neurotransmitters [Baseline, at the completion of therapy, and every 12 months up to 36 months after off therapy date]

    15. Change in Neurocognitive Performance [Baseline, at the completion of therapy, and every 12 months up to 60 months off therapy]

      Age standardized performance on measures of global cognitive functioning, attention, processing speed and executive functions.

    16. Number and Type of Genetic Polymorphisms [At study enrollment (Day 0)]

    17. Pharmacogenetic Variation on CNS Transmitters [At study enrollment (Day 0)]

    18. Change in Neuropsychological Performance [Baseline, 6- and 12-months from treatment initiation, and yearly after the first year (up to 5 years)]

      The primary interest is in global cognitive functioning. This is measured using the SB-V Routing subtests.

    19. Change in Quantitative Magnetic Resonance (MR) Measures in the Frontal Lobe [Baseline and up to 60 months after completion of therapy.]

    20. Change in Neurocognitive Performance in Attention, Working Memory, and Fluency [Baseline and prior to cycle A1 (~6 months) and at end of therapy and at 12, 24, 36, 48 and 60 months after completion of therapy.]

      Neurocognitive performance is assessed using a comprehensive battery of standard tests. Sustained attention is measured using the TOVA; selective auditory attention is measured using the WJIII; nonverbal attention span is measured using the SB-V Block Span subset. Working memory is measured using the WJIII. Fluency is measured using is also measured using the WJIII.

    21. Change in Quantitative MR Measures in the Right Frontal-parietal Regions [Baseline and up to 5 years after completion of therapy]

    22. Change in Neurocognitive Performance in Visual-spatial Reasoning and Processing Speed [Baseline and up to 5 years after completion of therapy.]

      Processing speed will be measured using the WJIII. Visual perception and visual-motor integration will be measured using the Beery VMI.

    23. Number of Participants With Endocrinopathy [Baseline, end of therapy, and at 6- and 24-months after completion of therapy]

      Serial GH testing (at baseline, the end of therapy, and at 6 and 24 months after completion of therapy) will be performed on consenting patients in order to estimate longitudinal change in GH secretion as measured by mean peak GH values, with the intent to explore associations with radiation dose to the hypothalamus. Since determination of proton- or photon-based radiotherapy is not based on randomization, it will not be possible to compare the endocrine outcome between the patients with and without PBT. However, the differences between these two clinical cohorts with respect to clinical and demographic variables of interest will be summarized via descriptive statistics.

    24. Longitudinal Change in Growth Hormone Secretion [Baseline, end of therapy, and at 6- and 24-months after completion of therapy]

      The intent of this objective is to estimate the longitudinal change in abnormal GH secretion as measured by mean peak GH values via a mixed effects model for the patients who receive PBT.

    25. Methotrexate Clearance in Induction Cycle 1 [Pre-infusion and 6, 23, 42, 66 hours from start of Methotrexate (MTX)]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.

    26. Methotrexate Clearance in Induction Cycle 2 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.

    27. Methotrexate Clearance in Induction Cycle 3 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.

    28. Methotrexate Clearance in Induction Cycle 4 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.

    29. Methotrexate Volume of Central Compartment in Induction Cycle 1 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.

    30. Methotrexate Volume of Central Compartment in Induction Cycle 2 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.

    31. Methotrexate Volume of Central Compartment in Induction Cycle 3 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.

    32. Methotrexate Volume of Central Compartment in Induction Cycle 4 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.

    33. Methotrexate AUC0-66h in Induction Cycle 1 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.

    34. Methotrexate AUC0-66h in Induction Cycle 2 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.

    35. Methotrexate AUC0-66h in Induction Cycle 3 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.

    36. Methotrexate AUC0-66h in Induction Cycle 4 [Pre-infusion and 6, 23, 42, 66 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.

    37. Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 1 [42 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.

    38. Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 2 [42 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.

    39. Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 3 [42 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.

    40. Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 4 [42 hours from start of MTX]

      Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.

    41. Cyclophosphamide Clearance in Induction Chemotherapy [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Cyclophosphamide plasma concentration-time data are collected on day 9 in one cycle of induction chemotherapy. Individual estimates of cyclophosphamide clearance are obtained using post hoc analysis.

    42. Cyclophosphamide Clearance in Consolidation Chemotherapy Cycle 1 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of cyclophosphamide clearance are obtained using post hoc analysis.

    43. Cyclophosphamide Clearance in Consolidation Chemotherapy Cycle 2 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of cyclophosphamide clearance are obtained using post hoc analysis.

    44. Cyclophosphamide Apparent Oral Clearance in Maintenance Chemotherapy Cycle A1 [Pre-dose, 0.5, 1.75, 3 and 6 hours post-dose]

      Cyclophosphamide plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of cyclophosphamide apparent oral clearance are obtained using post hoc analysis.

    45. Cyclophosphamide AUC0-24h in Induction Chemotherapy [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Cyclophosphamide plasma concentration-time data are collected on day 9 in one cycle of induction chemotherapy. Individual estimates of cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.

    46. Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 1 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.

    47. Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 2 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.

    48. Cyclophosphamide AUC0-24h in Maintenance Chemotherapy Cycle A1 [Pre-dose, 0.5, 1.75, 3, 6, and 24 hours post-dose]

      Cyclophosphamide plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of cyclophosphamide AUC0-24h are obtained using post hoc analysis.

    49. 4-OH Cyclophosphamide AUC0-24h in Induction Chemotherapy [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      4-OH cyclophosphamide plasma concentration-time data are collected on day 9 in one cycle of induction chemotherapy. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.

    50. 4-OH Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 1 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      4-OH cyclophosphamide plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.

    51. 4-OH Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 2 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      4-OH cyclophosphamide plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.

    52. 4-OH Cyclophosphamide AUC0-24h in Maintenance Chemotherapy Cycle A1 [Pre-dose, 0.5, 1.75, 3, 6, and 24 hours post-dose]

      4-OH cyclophosphamide plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.

    53. CEPM AUC0-24h in Induction Chemotherapy [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected on day 9 in one induction cycle. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.

    54. CEPM AUC0-24h in Consolidation Chemotherapy Cycle 1 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.

    55. CEPM AUC0-24h in Consolidation Chemotherapy Cycle 2 [Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion]

      Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.

    56. CEPM AUC0-24h in Maintenance Chemotherapy Cycle A1 [Pre-dose, 0.5, 1.75, 3, 6, and 24 hours post-dose]

      Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.

    57. Participants With Empirical Dosage Achieving Target System Exposure of Intravenous Topotecan [Pre-infusion, 5 min., 1, and 3 hours from end of infusion]

      Number of participants who successfully achieve target systemic exposure of intravenous topotecan after an empiric dosage during consolidation phase of therapy are reported.

    58. Participants With PK-guided Dosage Adjustment Achieving Target System Exposure of Intravenous Topotecan [Pre-infusion, 5 min., 1, and 3 hours from end of infusion]

      Number of participants who successfully achieve target systemic exposure of intravenous topotecan after a pharmacokinetic-guided dosage adjustment during consolidation phase of therapy are reported.

    59. Topotecan Clearance in Consolidation Chemotherapy [Pre-infusion, 5 min., 1, and 3 hours from end of infusion]

      Topotecan plasma concentration-time data are collected on day 1 of consolidation cycle 1 after a single IV dose. Individual estimates of topotecan clearance are obtained using post hoc analysis.

    60. Topotecan Apparent Oral Clearance in Maintenance Chemotherapy [Pre-dose, 0.25, 1.5 and 6 hours post-dose]

      Topotecan plasma concentration-time data are collected on day 1 of maintenance cycle A1 after a single oral dose. Individual estimates of topotecan apparent oral clearance are obtained using post hoc analysis.

    61. Topotecan AUC0-24h in Consolidation Chemotherapy [Pre-infusion, 5 min., 1, 3, and 24 hours from end of infusion]

      Topotecan plasma concentration-time data are collected on day 1 of consolidation cycle 1 after a single IV dose. Individual estimates of topotecan AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.

    62. Topotecan AUC0-24h in Maintenance Chemotherapy [Pre-dose, 0.25, 1.5, 6, and 24 hours post-dose]

      Topotecan plasma concentration-time data are collected on day 1 of maintenance cycle A1 after a single oral dose. Individual estimates of topotecan AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.

    63. Erlotinib Apparent Oral Clearance [Pre-dose, 1, 2, 4, 8, and 24 hours post-dose]

      Erlotinib plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of erlotinib apparent oral clearance are obtained using post hoc analysis.

    64. Erlotinib Apparent Volume of Central Compartment [Pre-dose, 1, 2, 4, 8, and 24 hours post-dose]

      Erlotinib plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of erlotinib apparent volume of central compartment are obtained using post hoc analysis.

    65. Erlotinib AUC0-24h [Pre-dose, 1, 2, 4, 8, and 24 hours post-dose]

      Erlotinib plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of erlotinib AUC0-24h (area under concentration curve from 0 to 24 hours post-dose) are obtained using post hoc analysis.

    66. OSI-420 AUC0-24h [Pre-dose, 1, 2, 4, 8, and 24 hours post-dose]

      Erlotinib metabolite OSI-420 plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of OSI-420 AUC0-24h (area under concentration curve from 0 to 24 hours post-dose) are obtained using post hoc analysis.

    67. Rate of Local Disease Progression [1 year after completion of radiation therapy for last patient]

      Local failure was defined as the interval from end of RT to date of local failure (or combined local + distant failure). Competing events were distant failure or second malignancy. Patients without an event were censored at date of last contact. The 1-year cumulative incidence was estimated and reported with a 95% confidence interval.

    68. Rate of Distant Disease Progression [1 year after completion of radiation therapy for last patient]

      Distant failure was defined as the interval from end of RT to date of distant failure (or combined local + distant failure). Competing events were local failure or second malignancy. Patients without an event were censored at date of last contact. The 1-year cumulative incidence was estimated and reported with a 95% confidence interval.

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    N/A to 5 Years
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Histologically confirmed newly diagnosed CNS tumors of any of the following :
    • Medulloblastoma (all histologic subtypes, including medullomyoblastoma and melanotic medulloblastoma)

    • Supratentorial primitive neuroectodermal tumor (PNET) (including CNS neuroblastoma or ganglioneuroblastoma, medulloepithelioma, and ependymoblastoma)

    • Pineoblastoma

    • Atypical teratoid rhabdoid tumor (ATRT)

    • Choroid plexus carcinoma

    • High grade glioma (including anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic ganglioglioma, pleomorphic xanthoastrocytoma with anaplastic features, high-grade astroblastoma , anaplastic pilocytic astrocytoma, malignant glioneuronal tumor, glioblastoma multiforme), or gliosarcoma,

    • Ependymoma (including all ependymoma histological variants)

    • Age < 3 years at time of diagnosis for all histological diagnosis. Medulloblastoma patients ≥ 3 and < 5years old at diagnosis who have non-metastatic disease with no more than 1cm2 of residual tumor are also eligible.

    • Meets criteria for 1 of the following risk groups:

    • Low-risk group:

    • Histologically confirmed nodular desmoplastic medulloblastoma, including medulloblastoma with extensive nodularity

    • Focal areas of anaplasia or other atypical features suggesting more aggressive phenotype in a tumor otherwise considered nodular desmoplastic should be treated on the intermediate-risk group, with final risk stratification at the discretion of principal investigator and study pathologist

    • No evidence of CNS metastasis 7 to 28 days after surgery by MRI and cytologic examination of lumbar cerebrospinal fluid (CSF)

    • Ventricular CSF from a shunt or Ommaya reservoir may be used to rule out M1 disease when lumbar puncture is medically contraindicated

    • Intermediate-risk group assignment when there is no other evidence of metastasis and CSF sampling is not possible

    • Gross total resection, defined as residual tumor or imaging abnormality (not definitive for residual tumor) with a size of < 1 cm2 confirmed on postoperative CT scan or MRI

    • Brain stem invasion by the tumor in the absence of imaging evidence of residual tumor (tumor size < 1 cm2) and otherwise meets criteria for the low-risk group, the patient will be classified as low-risk

    • Desmoplastic medulloblastoma patients who are ≥3 -<5 years of age will NOT be eligible for the low risk arm of the protocol.

    • Intermediate-risk group:

    • Histologically confirmed nodular desmoplastic medulloblastoma with less than gross total resection and no evidence of metastasis

    • Any eligible histologic diagnosis other than desmoplastic medulloblastoma with no evidence of CNS metastasis

    • Medulloblastoma patients who are ≥3 and < 5 yrs of age irrespective of histology and with no evidence of CNS metastasis

    • High-risk group:

    • Any eligible histologic diagnosis with evidence of CNS metastasis

    • Patients with extraneural metastasis are eligible for treatment on the high-risk group

    PATIENT CHARACTERISTICS:
    • Lansky performance status ≥ 30 (except for posterior fossa syndrome)

    • WBC > 2,000/mm3

    • Platelets > 50,000/mm3 (without support)

    • Hemoglobin > 8 g/dL (with or without support)

    • ANC > 500/mm3

    • Serum creatinine < 3 times upper limit of normal (ULN)

    • ALT < 5 times ULN

    • Total bilirubin < 3 times ULN

    PRIOR CONCURRENT THERAPY:
    • See Disease Characteristics

    • No more than 31 days since prior definitive surgery

    • No prior radiotherapy or chemotherapy other than corticosteroid therapy

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Lucile Packard Children's Hospital at Stanford University Medical Center Palo Alto California United States 94304
    2 Rady Children's Hospital San Diego California United States 92123
    3 Children's Hospitals and Clinics of Minnesota - St. Paul Saint Paul Minnesota United States 55102
    4 St. Jude Children's Research Hospital Memphis Tennessee United States 38105
    5 University of Texas Southwestern Medical Center at Dallas Dallas Texas United States 75390
    6 Lady Cilento Children's Hospital, Brisbane Brisbane Queensland Australia 4029

    Sponsors and Collaborators

    • St. Jude Children's Research Hospital
    • University of Florida
    • National Cancer Institute (NCI)
    • The Pew Charitable Trusts

    Investigators

    • Study Chair: Amar Gajjar, MD, St. Jude Children's Research Hospital

    Study Documents (Full-Text)

    More Information

    Additional Information:

    Publications

    None provided.
    Responsible Party:
    St. Jude Children's Research Hospital
    ClinicalTrials.gov Identifier:
    NCT00602667
    Other Study ID Numbers:
    • SJYC07
    • R01CA154619
    • NCI-2011-01193
    First Posted:
    Jan 28, 2008
    Last Update Posted:
    Dec 6, 2021
    Last Verified:
    Dec 1, 2021

    Study Results

    Participant Flow

    Recruitment Details 293 participants were enrolled between December 17, 2007 and April 19, 2017.
    Pre-assignment Detail Of the 293 participants enrolled, 3 were ineligible and removed from study, leaving 290 eligible patients. Eighty-one (81) of the 290 eligible patients enrolled had histologically confirmed medulloblastoma; medulloblastoma patients are the focus of the primary study objectives.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic (M0) medulloblastoma, nodular desmoplastic or high grade glioma histology will receive induction chemotherapy and low-risk therapy. Patients with M0 medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease, will receive induction chemotherapy and intermediate-risk therapy. Patients with central nervous system (CNS) metastatic disease will receive induction chemotherapy and high-risk therapy.
    Period Title: Overall Study
    STARTED 57 156 77
    COMPLETED 38 75 14
    NOT COMPLETED 19 81 63

    Baseline Characteristics

    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group Total
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic (M0) medulloblastoma, nodular desmoplastic or high grade glioma histology will receive induction chemotherapy and low-risk therapy. Patients with M0 medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease, will receive induction chemotherapy and intermediate-risk therapy. Patients with central nervous system (CNS) metastatic disease will receive induction chemotherapy and high-risk therapy. Total of all reporting groups
    Overall Participants 57 156 77 290
    Age (months) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [months]
    17.0
    (10.0)
    22.9
    (12.8)
    19.3
    (10.4)
    20.8
    (11.9)
    Age (months) [Median (Full Range) ]
    Median (Full Range) [months]
    16.1
    21.9
    20.0
    20.1
    Sex: Female, Male (Count of Participants)
    Female
    22
    38.6%
    66
    42.3%
    43
    55.8%
    131
    45.2%
    Male
    35
    61.4%
    90
    57.7%
    34
    44.2%
    159
    54.8%
    Race/Ethnicity, Customized (Count of Participants)
    Mexican/Chicano
    0
    0%
    5
    3.2%
    2
    2.6%
    7
    2.4%
    NOS Spanish, Hispanic, Latino
    7
    12.3%
    16
    10.3%
    7
    9.1%
    30
    10.3%
    Non Spanish speaking, Non Hispanic
    47
    82.5%
    120
    76.9%
    64
    83.1%
    231
    79.7%
    Puerto Rican
    1
    1.8%
    2
    1.3%
    1
    1.3%
    4
    1.4%
    South or Central American
    0
    0%
    2
    1.3%
    0
    0%
    2
    0.7%
    Unknown
    2
    3.5%
    11
    7.1%
    3
    3.9%
    16
    5.5%
    Race/Ethnicity, Customized (Count of Participants)
    American Indian/Alaskan/White
    1
    1.8%
    0
    0%
    0
    0%
    1
    0.3%
    American Indian/Alaskan Native
    0
    0%
    1
    0.6%
    0
    0%
    1
    0.3%
    Asian
    3
    5.3%
    6
    3.8%
    1
    1.3%
    10
    3.4%
    Asian and White
    0
    0%
    2
    1.3%
    2
    2.6%
    4
    1.4%
    Black
    6
    10.5%
    14
    9%
    10
    13%
    30
    10.3%
    Black and White
    0
    0%
    1
    0.6%
    0
    0%
    1
    0.3%
    Multiple Race (NOS)
    1
    1.8%
    7
    4.5%
    1
    1.3%
    9
    3.1%
    Other
    2
    3.5%
    4
    2.6%
    1
    1.3%
    7
    2.4%
    Pacific Islander
    1
    1.8%
    0
    0%
    0
    0%
    1
    0.3%
    Unknown
    2
    3.5%
    3
    1.9%
    1
    1.3%
    6
    2.1%
    White
    41
    71.9%
    118
    75.6%
    61
    79.2%
    220
    75.9%

    Outcome Measures

    1. Primary Outcome
    Title Percent Probability of Progression-free Survival (PFS) for Medulloblastoma Patients
    Description Progression was defined as 25% increase in the size of any measurable lesion; the appearance of a new lesion; or the conversion of negative cerebrospinal fluid (CSF) cytology to positive. Defined as the time interval from date on treatment until the date of first progression, medulloblastoma-related death or date of last contact for patients who have not experienced an event. All eligible medulloblastoma patients who received any methotrexate are included in this analysis.
    Time Frame From date on treatment until date of first progression or relapse or disease related death or date of last contact, estimated at 1 year after treatment

    Outcome Measure Data

    Analysis Population Description
    All eligible medulloblastoma patients started methotrexate and were included (n=81). PFS estimates are reported by risk group.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Medulloblastoma patients <3 years of age; histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN); no evidence of metastatic disease (M0); and a surgical gross total resection (GTR) or near total resection (NTR) defined as residual tumor or imaging abnormality (not definite for residual tumor) with a size of less than 1 cm^2 on post-op computed tomography (CT) or magnetic resonance imaging (MRI) [R0]) Medulloblastoma patients <3 years of age, M0, and either R0 resection with any other medullo histology other than DN/MBEN (i.e., classic or large cell anaplastic (LCA)), or with a subtotal resection (with a residual tumor or imaging abnormality with a size of >1 cm^2 on post-op imaging (R+), and DN/MBEN). Children 3-5 years of age were also included in this group High Risk Medulloblastoma patients <3 years of age with evidence of metastatic disease
    Measure Participants 23 32 26
    Number (95% Confidence Interval) [Percent Probability]
    73.9
    46.9
    30.8
    2. Primary Outcome
    Title Percent Probability of Progression-free Survival (PFS) for Medulloblastoma Patients by DNA Methylation Subgroup
    Description Defined as the time interval from date on treatment until the date of first progression, medulloblastoma-related death or date of last contact for patients who have not experienced an event. Eligible medulloblastoma patients who received any methotrexate and had molecularly confirmed medulloblastoma are included in this analysis. Five patients were excluded as 3 had no archival tissue available and 2 were found to not be medulloblastoma by methylation profile.
    Time Frame From date on treatment to date of first progression or relapse or disease related death or date of last contact, estimated at 1 year after treatment

    Outcome Measure Data

    Analysis Population Description
    Eligible patients with molecularly confirmed medulloblastoma (n=76) were included. PFS estimates are reported by methylation subgroup and risk group. There were no low-risk group 3 or group 4 patients.
    Arm/Group Title Low-risk SHH Patients Intermediate-risk SHH Patients High-risk SHH Patients Low-risk Group 3 Patients Intermediate-risk Group 3 Patients High-risk Group 3 Patients Low-risk Group 4 Patients Intermediate-risk Group 4 Patients High-risk Group 4 Patients
    Arm/Group Description Sonic hedgehog (SHH) medulloblastoma subgroup patients in the low-risk group Sonic hedgehog (SHH) medulloblastoma subgroup patients in the intermediate-risk group Sonic hedgehog (SHH) medulloblastoma subgroup patients in the high-risk group Group 3 (G3) medulloblastoma subgroup patients in the low-risk group Group 3 medulloblastoma subgroup patients in the intermediate-risk group Group 3 medulloblastoma subgroup patients in the high-risk group Group 4 (G4) medulloblastoma subgroup patients in the low-risk group Group 4 (G4) medulloblastoma subgroup patients in the intermediate-risk group Group 4 medulloblastoma subgroup patients in the high-risk group
    Measure Participants 23 8 11 0 13 11 0 8 2
    Number (95% Confidence Interval) [Percent Probability]
    73.9
    50.0
    54.5
    30.8
    9.1
    62.5
    50.0
    3. Primary Outcome
    Title Percent Probability of Event-free Survival (EFS) for Medulloblastoma Patients
    Description Defined as the time interval from date on treatment until the date of first progression, second malignancy or death due to any cause; or date of last contact for patients who have not experienced an event. All eligible medulloblastoma patients who received any methotrexate are included in this analysis.
    Time Frame From date on treatment to date of first progression, relapse, second malignancy or death from any cause or to date of last contact, estimated at 1 year after

    Outcome Measure Data

    Analysis Population Description
    Eligible medulloblastoma patients (n=81) were included. EFS estimates are reported by risk group.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Medulloblastoma patients <3 years of age; histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN); no evidence of metastatic disease (M0); and a surgical gross total resection (GTR) or near total resection (NTR) defined as residual tumor or imaging abnormality (not definite for residual tumor) with a size of less than 1 cm^2 on post-op computed tomography (CT) or magnetic resonance imaging (MRI) [R0]) Medulloblastoma patients <3 years of age, M0, and either R0 resection with any other medullo histology other than DN/MBEN (i.e., classic or large cell anaplastic (LCA)), or with a subtotal resection (with a residual tumor or imaging abnormality with a size of >1 cm^2 on post-op imaging (R+), and DN/MBEN). Children 3-5 years of age were also included in this group High Risk Medulloblastoma patients <3 years of age with evidence of metastatic disease
    Measure Participants 23 32 26
    Number (95% Confidence Interval) [Percent Probability]
    73.9
    46.9
    30.8
    4. Secondary Outcome
    Title Number of Participants With Chromosomal Abnormalities
    Description Amplifications and deletions (gains and losses) for chromosomes of interest are shown in the table of measured values.
    Time Frame Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment

    Outcome Measure Data

    Analysis Population Description
    Eligible medulloblastoma patients (n=81) were included in these analyses. 23 of 23 low risk patients had data available for this objective, as did 27/32 and 24/26 intermediate and high risk patients.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Medulloblastoma patients <3 years of age; histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN); no evidence of metastatic disease (M0); and a surgical gross total resection (GTR) or near total resection (NTR) defined as residual tumor or imaging abnormality (not definite for residual tumor) with a size of less than 1 cm^2 on post-op computed tomography (CT) or magnetic resonance imaging (MRI) [R0]) Medulloblastoma patients <3 years of age, M0, and either R0 resection with any other medullo histology other than DN/MBEN (i.e., classic or large cell anaplastic (LCA)), or with a subtotal resection (with a residual tumor or imaging abnormality with a size of >1 cm^2 on post-op imaging (R+), and DN/MBEN). Children 3-5 years of age were also included in this group High Risk Medulloblastoma patients <3 years of age with evidence of metastatic disease
    Measure Participants 23 32 26
    chr2p gain/amplification
    5
    8.8%
    1
    0.6%
    6
    7.8%
    chr2p loss/deletion
    0
    0%
    2
    1.3%
    0
    0%
    chr2q gain/amplification
    5
    8.8%
    1
    0.6%
    6
    7.8%
    chr2q loss/deletion
    0
    0%
    2
    1.3%
    0
    0%
    chr6p gain/amplification
    1
    1.8%
    3
    1.9%
    3
    3.9%
    chr6p loss/deletion
    0
    0%
    0
    0%
    2
    2.6%
    chr6q gain/amplification
    1
    1.8%
    3
    1.9%
    3
    3.9%
    chr6q loss/deletion
    0
    0%
    0
    0%
    2
    2.6%
    chr8p gain/amplification
    1
    1.8%
    2
    1.3%
    0
    0%
    chr8p loss/deletion
    0
    0%
    6
    3.8%
    8
    10.4%
    chr8q gain/amplification
    1
    1.8%
    3
    1.9%
    0
    0%
    chr8q loss/deletion
    0
    0%
    5
    3.2%
    7
    9.1%
    chr9p gain/amplification
    4
    7%
    6
    3.8%
    3
    3.9%
    chr9p loss/deletion
    4
    7%
    0
    0%
    3
    3.9%
    chr9q gain/amplification
    2
    3.5%
    1
    0.6%
    1
    1.3%
    chr9q loss/deletion
    6
    10.5%
    5
    3.2%
    5
    6.5%
    chr10p gain/amplification
    0
    0%
    0
    0%
    0
    0%
    chr10p loss/deletion
    0
    0%
    5
    3.2%
    7
    9.1%
    chr10q gain/amplification
    0
    0%
    0
    0%
    0
    0%
    chr10q loss/deletion
    3
    5.3%
    7
    4.5%
    9
    11.7%
    chr20p gain/amplification
    0
    0%
    0
    0%
    0
    0%
    chr20p loss/deletion
    2
    3.5%
    4
    2.6%
    5
    6.5%
    chr20q gain/amplification
    0
    0%
    0
    0%
    0
    0%
    chr20q loss/deletion
    1
    1.8%
    3
    1.9%
    5
    6.5%
    5. Secondary Outcome
    Title Numbers of Patients With Gene Alterations
    Description Gene alterations, which include single nucleotide variants (SNPs), amplifications, deletions, translocations, indels, and germline alterations are shown for specific genes of interest in the results table.
    Time Frame Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment

    Outcome Measure Data

    Analysis Population Description
    Eligible medulloblastoma patients (n=81) were included in these analyses. Not all patients had data available for each gene.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Medulloblastoma patients <3 years of age; histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN); no evidence of metastatic disease (M0); and a surgical gross total resection (GTR) or near total resection (NTR) defined as residual tumor or imaging abnormality (not definite for residual tumor) with a size of less than 1 cm^2 on post-op computed tomography (CT) or magnetic resonance imaging (MRI) [R0]) Medulloblastoma patients <3 years of age, M0, and either R0 resection with any other medullo histology other than DN/MBEN (i.e., classic or large cell anaplastic (LCA)), or with a subtotal resection (with a residual tumor or imaging abnormality with a size of >1 cm^2 on post-op imaging (R+), and DN/MBEN). Children 3-5 years of age were also included in this group High Risk Medulloblastoma patients <3 years of age with evidence of metastatic disease
    Measure Participants 23 32 26
    PTCH1 alteration
    7
    12.3%
    4
    2.6%
    3
    3.9%
    SUFU alteration
    6
    10.5%
    1
    0.6%
    1
    1.3%
    KMT2D alteration
    5
    8.8%
    1
    0.6%
    3
    3.9%
    SMO alteration
    2
    3.5%
    0
    0%
    1
    1.3%
    BCOR alteration
    1
    1.8%
    0
    0%
    0
    0%
    PTEN alteration
    1
    1.8%
    0
    0%
    0
    0%
    BRCA2 alteration
    0
    0%
    0
    0%
    0
    0%
    GLI2 alteration
    0
    0%
    0
    0%
    0
    0%
    SMARCA4 alteration
    2
    3.5%
    1
    0.6%
    0
    0%
    TP53 alteration
    0
    0%
    0
    0%
    0
    0%
    MYCN alteration
    0
    0%
    1
    0.6%
    1
    1.3%
    6. Secondary Outcome
    Title Numbers of Patients With Molecular Abnormalities by Tumor Type
    Description Alterations included single nucleotide variants (SNPs), amplifications, deletions, translocations, indels, and germline alterations. Cytogenetic information shows gains and losses as specified in the table of measured values.
    Time Frame Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment

    Outcome Measure Data

    Analysis Population Description
    Eligible patients with molecularly confirmed medulloblastoma (n=76) were included in these analyses. Not all patients had data available for each gene.
    Arm/Group Title Low-risk SHH Patients Intermediate-risk SHH Patients High-risk SHH Patients Low-risk Group 3 Patients Intermediate-risk Group 3 Patients High-risk Group 3 Patients Low-risk Group 4 Patients Intermediate-risk Group 4 Patients High-risk Group 4 Patients
    Arm/Group Description Sonic hedgehog (SHH) medulloblastoma subgroup patients in the low-risk group Sonic hedgehog (SHH) medulloblastoma subgroup patients in the intermediate-risk group Sonic hedgehog (SHH) medulloblastoma subgroup patients in the high-risk group Group 3 (G3) medulloblastoma subgroup patients in the low-risk group Group 3 medulloblastoma subgroup patients in the intermediate-risk group Group 3 medulloblastoma subgroup patients in the high-risk group Group 4 (G4) medulloblastoma subgroup patients in the low-risk group Group 4 (G4) medulloblastoma subgroup patients in the intermediate-risk group Group 4 medulloblastoma subgroup patients in the high-risk group
    Measure Participants 23 8 11 0 13 11 0 8 2
    PTCH1 alteration
    7
    12.3%
    4
    2.6%
    3
    3.9%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    SUFU alteration
    6
    10.5%
    1
    0.6%
    1
    1.3%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    KMT2D alteration
    5
    8.8%
    1
    0.6%
    3
    3.9%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    SMO alteration
    2
    3.5%
    0
    0%
    1
    1.3%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    BCOR alteration
    1
    1.8%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    PTEN alteration
    1
    1.8%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    BRCA2 alteration
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    GLI2 alteration
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    SMARCA4 alteration
    2
    3.5%
    0
    0%
    0
    0%
    1
    0.3%
    0
    NaN
    0
    NaN
    0
    NaN
    TP53 alteration
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    MYCN amplification
    0
    0%
    1
    0.6%
    0
    0%
    0
    0%
    1
    NaN
    0
    NaN
    0
    NaN
    chr2p gain/amplification
    5
    8.8%
    0
    0%
    4
    5.2%
    0
    0%
    1
    NaN
    0
    NaN
    0
    NaN
    chr2p loss/deletion
    0
    0%
    0
    0%
    0
    0%
    1
    0.3%
    0
    NaN
    1
    NaN
    0
    NaN
    chr2q gain/amplification
    5
    8.8%
    0
    0%
    4
    5.2%
    0
    0%
    1
    NaN
    0
    NaN
    0
    NaN
    chr2q loss/deletion
    0
    0%
    0
    0%
    0
    0%
    1
    0.3%
    0
    NaN
    1
    NaN
    0
    NaN
    chr6p gain/amplification
    1
    1.8%
    0
    0%
    0
    0%
    2
    0.7%
    3
    NaN
    1
    NaN
    0
    NaN
    chr6p loss/deletion
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    2
    NaN
    0
    NaN
    0
    NaN
    chr6q gain/amplification
    1
    1.8%
    0
    0%
    0
    0%
    2
    0.7%
    3
    NaN
    1
    NaN
    0
    NaN
    chr6q loss/deletion
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    2
    NaN
    0
    NaN
    0
    NaN
    chr8p gain/amplification
    1
    1.8%
    0
    0%
    0
    0%
    1
    0.3%
    0
    NaN
    0
    NaN
    0
    NaN
    chr8p loss/deletion
    0
    0%
    0
    0%
    0
    0%
    2
    0.7%
    7
    NaN
    4
    NaN
    1
    NaN
    chr8q gain/amplification
    1
    1.8%
    0
    0%
    0
    0%
    2
    0.7%
    0
    NaN
    0
    NaN
    0
    NaN
    chr8q loss/deletion
    0
    0%
    0
    0%
    0
    0%
    1
    0.3%
    6
    NaN
    4
    NaN
    1
    NaN
    chr9p gain/amplification
    4
    7%
    4
    2.6%
    3
    3.9%
    1
    0.3%
    0
    NaN
    1
    NaN
    0
    NaN
    chr9p loss/deletion
    4
    7%
    0
    0%
    0
    0%
    0
    0%
    3
    NaN
    0
    NaN
    0
    NaN
    chr9q gain/amplification
    2
    3.5%
    0
    0%
    1
    1.3%
    0
    0%
    0
    NaN
    1
    NaN
    0
    NaN
    chr9q loss/deletion
    6
    10.5%
    5
    3.2%
    2
    2.6%
    0
    0%
    3
    NaN
    0
    NaN
    0
    NaN
    chr10p gain/amplification
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    chr10p loss/deletion
    0
    0%
    0
    0%
    0
    0%
    5
    1.7%
    7
    NaN
    0
    NaN
    0
    NaN
    chr10q gain/amplification
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    chr10q loss/deletion
    3
    5.3%
    2
    1.3%
    1
    1.3%
    5
    1.7%
    8
    NaN
    0
    NaN
    0
    NaN
    chr20p gain/amplification
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    chr20p loss/deletion
    2
    3.5%
    1
    0.6%
    1
    1.3%
    3
    1%
    4
    NaN
    0
    NaN
    0
    NaN
    chr20q gain/amplification
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    NaN
    0
    NaN
    0
    NaN
    chr20q loss/deletion
    1
    1.8%
    0
    0%
    1
    1.3%
    3
    1%
    4
    NaN
    0
    NaN
    0
    NaN
    7. Secondary Outcome
    Title Number of Successful Collections for Frozen and Fixed Tumor Samples
    Description Successful collections will be defined as the number of patients who have frozen/fixed tumor samples available.
    Time Frame Based on samples obtained at the time of initial surgery or repeat surgery prior to treatment

    Outcome Measure Data

    Analysis Population Description
    All eligible patients enrolled (n=290) were included in this analysis. The numbers of patients with pre-study samples were considered for these results.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic or high grade glioma histology will receive induction chemotherapy and low-risk therapy. Patients with (M0) medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease, will receive induction chemotherapy and intermediate-risk therapy. Patients with central nervous system (CNS) metastatic disease will receive induction chemotherapy and high-risk therapy.
    Measure Participants 57 156 77
    Number with frozen tumor tissue
    27
    47.4%
    73
    46.8%
    32
    41.6%
    Number with fixed tumor tissue
    54
    94.7%
    153
    98.1%
    71
    92.2%
    8. Secondary Outcome
    Title Event-free Survival (EFS) Compared to Historical Controls
    Description EFS was measured from the date of initial treatment to the earliest date of disease progression, second malignancy or death for patients who fail; and to the date of last contact for patients who remain at risk for failure. 1-year EFS estimates are reported by risk group. EFS was compared to St. Jude historical cohorts by risk group using hazard ratios with 95% confidence intervals.
    Time Frame From date on treatment until date of first event (progression, second malignancy or death) or until date of last contact, assessed up to 10 years

    Outcome Measure Data

    Analysis Population Description
    Eligible medulloblastoma patients who received any methotrexate were included in this analysis. Hazard ratios with 95% confidence intervals are reported and compare SJYC07 patients to historical controls in each risk group. As there were too few low-risk historical controls, no hazard ratio is included for the low-risk group.
    Arm/Group Title SJYC07 Low-risk Medulloblastoma Patients SJYC07 Intermediate-risk Medulloblastoma Patients SJYC07 High-risk Medulloblastoma Patients
    Arm/Group Description Medulloblastoma patients <3 years of age; histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN); no evidence of metastatic disease (M0); and a surgical gross total resection (GTR) or near total resection (NTR) defined as residual tumor or imaging abnormality (not definite for residual tumor) with a size of less than 1 cm^2 on post-op computed tomography (CT) or magnetic resonance imaging (MRI) [R0]) SJYC07 medulloblastoma patients <3 years of age, no evidence of metastatic disease (M0), and either R0 resection with any other medullo histology other than histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN) (i.e., classic or large cell anaplastic (LCA)), or with a subtotal resection (with a residual tumor or imaging abnormality with a size of >1 cm^2 on post-op imaging (R+), and DN/MBEN). Children 3-5 years of age were also included in this group SJYC07 medulloblastoma patients <3 years of age with evidence of metastatic disease
    Measure Participants 23 32 26
    Number (95% Confidence Interval) [Percent probability]
    73.9
    46.9
    30.8
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Intermediate-Risk Group
    Comments The historical control included 10 medulloblastoma patients treated during 1998-2006 who would have been classified as intermediate risk according to SJYC07 criteria (section 13.1.3 of protocol).
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 4.99
    Confidence Interval (2-Sided) 95%
    1.17 to 21.23
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection High-Risk Group
    Comments The historical control included 14 medulloblastoma patients treated during 1998-2006 who would have been classified as high risk according to SJYC07 criteria (section 13.1.3 of protocol).
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.86
    Confidence Interval (2-Sided) 95%
    0.40 to 1.84
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    9. Secondary Outcome
    Title Overall Survival (OS) Compared to Historical Controls
    Description OS was measured from the date of initial treatment to date of death or to date of last contact for survivors. 1-year OS estimates were reported by risk group. OS was compared to St. Jude historical cohorts by risk group using hazard ratios with 95% confidence intervals.
    Time Frame 1 year after treatment initiation of last patient

    Outcome Measure Data

    Analysis Population Description
    Eligible medulloblastoma patients who received any methotrexate were included in this analysis. Hazard ratios with 95% confidence intervals are reported and compare SJYC07 patients to historical controls in each risk group. As there were too few low-risk historical controls, no hazard ratio is included for the low-risk group.
    Arm/Group Title SJYC07 Low-risk Medulloblastoma Patients SJYC07 Intermediate-risk Medulloblastoma Patients SJYC07 High-risk Medulloblastoma Patients
    Arm/Group Description Medulloblastoma patients <3 years of age; histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN); no evidence of metastatic disease (M0); and a surgical gross total resection (GTR) or near total resection (NTR) defined as residual tumor or imaging abnormality (not definite for residual tumor) with a size of less than 1 cm^2 on post-op computed tomography (CT) or magnetic resonance imaging (MRI) [R0]) SJYC07 medulloblastoma patients <3 years of age, no evidence of metastatic disease (M0), and either R0 resection with any other medullo histology other than histological diagnosis of desmoplastic nodular (DN)/medulloblastoma with extensive nodularity (MBEN) (i.e., classic or large cell anaplastic (LCA)), or with a subtotal resection (with a residual tumor or imaging abnormality with a size of >1 cm^2 on post-op imaging (R+), and DN/MBEN). Children 3-5 years of age were also included in this group SJYC07 medulloblastoma patients <3 years of age with evidence of metastatic disease
    Measure Participants 23 32 26
    Number (95% Confidence Interval) [Percent probability]
    100
    84.4
    61.5
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Intermediate-Risk Group
    Comments The historical control included 10 medulloblastoma patients treated during 1998-2006 who would have been classified as intermediate risk according to SJYC07 criteria (section 13.1.3 of protocol).
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 1.85
    Confidence Interval (2-Sided) 95%
    0.42 to 8.22
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection High-Risk Group
    Comments The historical control included 14 medulloblastoma patients treated during 1998-2006 who would have been classified as high risk according to SJYC07 criteria (section 13.1.3 of protocol).
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.75
    Confidence Interval (2-Sided) 95%
    0.31 to 1.79
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    10. Secondary Outcome
    Title Percentage of Patients With Objective Responses Rate to Induction Chemotherapy
    Description For patients treated in the intermediate and high risk strata with residual or metastatic disease we will estimate the stratum-specific objective response rate (complete response (CR) or partial response [ PR]). All patients who receive at least 1 -dose of methotrexate are evaluable for response. Objective responses must be sustained for at least eight weeks.
    Time Frame From on-study date to 2 months after completion of induction chemotherapy (up to 4 months after on-study date)

    Outcome Measure Data

    Analysis Population Description
    Eligible intermediate and high risk group patients who received at least 1 dose of methotrexate were included in this analysis. One of the high risk patients did not start therapy and was excluded.
    Arm/Group Title Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 156 76
    Number (95% Confidence Interval) [Percentage of patients]
    58.3
    21.1
    11. Secondary Outcome
    Title Feasibility and Toxicity of Administering Vinblastine With Induction Chemotherapy for Patients With Metastatic Disease as Measured by the Percentage of Courses Delayed for More Than 7 Days Due to Toxicity
    Description For the subset of patients with metastatic disease (high-risk group patients), during induction, the proportion percentage of courses during which subsequent chemotherapy administration was delayed for more than 7 days due to toxicity will be calculated. Patients were to receive 4 courses of induction and then consolidation chemotherapy.
    Time Frame From on-study date up to 4 months after on-study date

    Outcome Measure Data

    Analysis Population Description
    Eligible high-risk group patients who started therapy were included in this analysis (n=76). 1 patient enrolled on the high-risk arm did not start therapy due to early disease progression and was excluded. Courses 2-4 of induction and the 1st consolidation course were used in this analysis. 76 patients (with 263 courses) were included.
    Arm/Group Title High-Risk Group
    Arm/Group Description Patients with central nervous system (CNS) metastatic disease
    Measure Participants 76
    Measure courses 263
    Number (95% Confidence Interval) [Percentage of courses delayed]
    3.8
    12. Secondary Outcome
    Title Feasibility and Toxicity of Administering Consolidation Therapy Including Cyclophosphamide and Pharmacokinetically Targeted Topotecan to Patients With Metastatic Disease Based on the Percentage of Courses Delayed for More Than 7 Days Due to Toxicity
    Description For the subset of patients with metastatic disease (high-risk group patients), during consolidation, we will calculate the number and proportion of courses during which subsequent chemotherapy administration was delayed for more than 7 days due to toxicity. Patients were to received 2 courses of consolidation chemotherapy and then maintenance therapy.
    Time Frame At completion of consolidation therapy (up to 6 months after on-study date)

    Outcome Measure Data

    Analysis Population Description
    Eligible high-risk group patients who started therapy were included. Course 2 of consolidation and the 1st course of maintenance were used in this analysis and assessed for delays >7 days due to toxicity. Of the 76 high-risk patients that started induction therapy, 50 started consolidation (with 47 courses included in this analysis).
    Arm/Group Title High-Risk Group
    Arm/Group Description Patients with central nervous system (CNS) metastatic disease
    Measure Participants 50
    Measure courses 47
    Number (95% Confidence Interval) [Percentage of courses delayed]
    2.6
    13. Secondary Outcome
    Title Percent of Patients With Sustained Objective Responses Rate After Consolidation
    Description For patients enrolled on the high-risk arm with measurable residual disease after induction treated with consolidation therapy, we will estimate the objective response (complete response (CR)/partial response (PR)) rate after consolidation therapy with a 95% confidence interval. Objective responses must be sustained for at least eight weeks. All patients who receive at least 1 dose of cyclophosphamide or topotecan during consolidation are evaluable for response.
    Time Frame 8 weeks after completion of consolidation therapy (up to 8 months after on-study date)

    Outcome Measure Data

    Analysis Population Description
    Of the 50 high-risk patients that started consolidation chemotherapy, 38 had measurable residual disease after induction and were included in this result.
    Arm/Group Title High-Risk Group
    Arm/Group Description Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38
    Number (95% Confidence Interval) [percentage of participants]
    13.2
    23.2%
    14. Secondary Outcome
    Title Feasibility and Toxicity of Administering Oral Maintenance Therapy in Children <3 Years of Age as Measured by the Percentage of Total Scheduled Maintenance Doses Received
    Description These data are based on patient diaries. For children <3 years of age, we will calculate the percentage of total scheduled doses each patient received per course for each of the oral maintenance courses and report the overall average number percentage of doses received per course across patients. If patients received all planned doses, their percentage would be 100%. If the average percentage was less than 75%, then feasibility would be in question.
    Time Frame From start of oral maintenance therapy (approximately 6 months after on-study date) to completion of oral maintenance therapy (up to 1 year after on-study date)

    Outcome Measure Data

    Analysis Population Description
    Eligible patients less than 3 years of age (n=273) constituted the study population for this analysis; 167 of these patients started the first course of maintenance and were included in this analysis (50 low-risk, 87 intermediate-risk, and 30 high-risk patients).
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients <3 years of age with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic or high grade glioma histology will receive induction chemotherapy and low-risk therapy. Patients <3 years of age with M0 medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease, will receive induction chemotherapy and intermediate-risk therapy. Patients <3 years of age with central nervous system (CNS) metastatic disease will receive induction chemotherapy and high-risk therapy
    Measure Participants 50 87 30
    Mean (Standard Deviation) [Percentage of scheduled doses received]
    96
    (8)
    91
    (23)
    98
    (4)
    15. Secondary Outcome
    Title Change in Neurostructure, Especially White Matter Volume and Integrity
    Description Quantitative MRI measures of change in neurostructure (especially white matter volume and integrity) over time will be assessed using a random effects model incorporating various covariates. Covariates to be considered include age at diagnosis, time since diagnosis and risk-arm. Differences in quantitative MRI measures of neurostructure volume and integrity between patient groups will be evaluated as a metric of structural neurotoxicity of therapy.
    Time Frame From baseline to 60 months off therapy

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    16. Secondary Outcome
    Title Percent of PET Scans With Loss of Signal Intensity
    Description Measures will be analyzed for intermediate risk participants who receive proton beam therapy (PBT) and who consent. This objective aims to assess the feasibility of using post-proton beam therapy (PBT) positron emission tomography (PET) as an in-vivo dosimetric and distal edge verification system in this patient population. To quantify the decay in signal, 134 scans from 53 patients were analyzed by recording the mean activation value (MAV), the average recorded PET signal from activation, within the target volume. With each patient being given the same dose, the percent standard deviation in the MAV can serve as a quantitative representation of signal loss due to radioactive decay.
    Time Frame Up to 3 times during RT consolidation

    Outcome Measure Data

    Analysis Population Description
    Intermediate risk patients treated at St. Jude were eligible to receive proton beam therapy through referral to the University of Florida Proton Therapy Institute. Patients electing to receive PBT and post-treatment PET scans after the delivery of one treatment beam were included in the analysis.
    Arm/Group Title Intermediate Risk Group
    Arm/Group Description Patients with M0 medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease
    Measure Participants 53
    Measure Scans 134
    Mean (Standard Deviation) [mean activation value (MAV)]
    60
    (27)
    17. Secondary Outcome
    Title Change in Concentration of Cerebrospinal Fluid (CSF) Neurotransmitters
    Description
    Time Frame Baseline, at the completion of therapy, and every 12 months up to 36 months after off therapy date

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    18. Secondary Outcome
    Title Change in Neurocognitive Performance
    Description Age standardized performance on measures of global cognitive functioning, attention, processing speed and executive functions.
    Time Frame Baseline, at the completion of therapy, and every 12 months up to 60 months off therapy

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    19. Secondary Outcome
    Title Number and Type of Genetic Polymorphisms
    Description
    Time Frame At study enrollment (Day 0)

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    20. Secondary Outcome
    Title Pharmacogenetic Variation on CNS Transmitters
    Description
    Time Frame At study enrollment (Day 0)

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    21. Secondary Outcome
    Title Change in Neuropsychological Performance
    Description The primary interest is in global cognitive functioning. This is measured using the SB-V Routing subtests.
    Time Frame Baseline, 6- and 12-months from treatment initiation, and yearly after the first year (up to 5 years)

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    22. Secondary Outcome
    Title Change in Quantitative Magnetic Resonance (MR) Measures in the Frontal Lobe
    Description
    Time Frame Baseline and up to 60 months after completion of therapy.

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    23. Secondary Outcome
    Title Change in Neurocognitive Performance in Attention, Working Memory, and Fluency
    Description Neurocognitive performance is assessed using a comprehensive battery of standard tests. Sustained attention is measured using the TOVA; selective auditory attention is measured using the WJIII; nonverbal attention span is measured using the SB-V Block Span subset. Working memory is measured using the WJIII. Fluency is measured using is also measured using the WJIII.
    Time Frame Baseline and prior to cycle A1 (~6 months) and at end of therapy and at 12, 24, 36, 48 and 60 months after completion of therapy.

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    24. Secondary Outcome
    Title Change in Quantitative MR Measures in the Right Frontal-parietal Regions
    Description
    Time Frame Baseline and up to 5 years after completion of therapy

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    25. Secondary Outcome
    Title Change in Neurocognitive Performance in Visual-spatial Reasoning and Processing Speed
    Description Processing speed will be measured using the WJIII. Visual perception and visual-motor integration will be measured using the Beery VMI.
    Time Frame Baseline and up to 5 years after completion of therapy.

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    26. Secondary Outcome
    Title Number of Participants With Endocrinopathy
    Description Serial GH testing (at baseline, the end of therapy, and at 6 and 24 months after completion of therapy) will be performed on consenting patients in order to estimate longitudinal change in GH secretion as measured by mean peak GH values, with the intent to explore associations with radiation dose to the hypothalamus. Since determination of proton- or photon-based radiotherapy is not based on randomization, it will not be possible to compare the endocrine outcome between the patients with and without PBT. However, the differences between these two clinical cohorts with respect to clinical and demographic variables of interest will be summarized via descriptive statistics.
    Time Frame Baseline, end of therapy, and at 6- and 24-months after completion of therapy

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    27. Secondary Outcome
    Title Longitudinal Change in Growth Hormone Secretion
    Description The intent of this objective is to estimate the longitudinal change in abnormal GH secretion as measured by mean peak GH values via a mixed effects model for the patients who receive PBT.
    Time Frame Baseline, end of therapy, and at 6- and 24-months after completion of therapy

    Outcome Measure Data

    Analysis Population Description
    [Not Specified]
    Arm/Group Title
    Arm/Group Description
    28. Secondary Outcome
    Title Methotrexate Clearance in Induction Cycle 1
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of Methotrexate (MTX)

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 1 and had samples collected for PK analysis
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 37 94 44
    Median (Full Range) [L/h/m^2]
    5.69
    6.06
    5.65
    29. Secondary Outcome
    Title Methotrexate Clearance in Induction Cycle 2
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 93 41
    Median (Full Range) [L/h/m^2]
    5.47
    5.70
    5.70
    30. Secondary Outcome
    Title Methotrexate Clearance in Induction Cycle 3
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 3 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 35 82 36
    Median (Full Range) [L/h/m^2]
    5.68
    5.78
    5.81
    31. Secondary Outcome
    Title Methotrexate Clearance in Induction Cycle 4
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 4 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 34 80 34
    Median (Full Range) [L/h/m^2]
    5.75
    5.89
    5.79
    32. Secondary Outcome
    Title Methotrexate Volume of Central Compartment in Induction Cycle 1
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 37 94 44
    Median (Full Range) [L/m^2]
    11.63
    13.70
    13.25
    33. Secondary Outcome
    Title Methotrexate Volume of Central Compartment in Induction Cycle 2
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 93 41
    Median (Full Range) [L/m^2]
    13.77
    13.73
    13.62
    34. Secondary Outcome
    Title Methotrexate Volume of Central Compartment in Induction Cycle 3
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 3 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 35 82 36
    Median (Full Range) [L/m^2]
    12.70
    13.55
    13.87
    35. Secondary Outcome
    Title Methotrexate Volume of Central Compartment in Induction Cycle 4
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate volume of central compartment are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 4 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 34 80 34
    Median (Full Range) [L/m^2]
    12.64
    13.31
    13.68
    36. Secondary Outcome
    Title Methotrexate AUC0-66h in Induction Cycle 1
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 37 94 44
    Median (Full Range) [µmol·h/L]
    1797
    1813
    1821
    37. Secondary Outcome
    Title Methotrexate AUC0-66h in Induction Cycle 2
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 93 41
    Median (Full Range) [µmol·h/L]
    1900
    1902
    1879
    38. Secondary Outcome
    Title Methotrexate AUC0-66h in Induction Cycle 3
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 3 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 35 82 36
    Median (Full Range) [µmol·h/L]
    1872
    1879
    1831
    39. Secondary Outcome
    Title Methotrexate AUC0-66h in Induction Cycle 4
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate AUC0-66h (area under concentration curve from time 0 to 66 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion and 6, 23, 42, 66 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 4 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 34 80 34
    Median (Full Range) [µmol·h/L]
    1804
    1841
    1886
    40. Secondary Outcome
    Title Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 1
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 1. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.
    Time Frame 42 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 37 93 44
    Median (Full Range) [µmol/L]
    0.49
    0.57
    0.61
    41. Secondary Outcome
    Title Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 2
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 2. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.
    Time Frame 42 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 93 41
    Median (Full Range) [µmol/L]
    0.75
    0.72
    0.69
    42. Secondary Outcome
    Title Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 3
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 3. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.
    Time Frame 42 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 3 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 35 82 36
    Median (Full Range) [µmol/L]
    0.65
    0.70
    0.58
    43. Secondary Outcome
    Title Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 4
    Description Methotrexate plasma concentration-time data are collected after the start of methotrexate infusion in induction cycle 4. Population parameters and inter-subject variability are estimated. Individual estimates of methotrexate concentration at 42 hours post-dose are obtained using post hoc analysis.
    Time Frame 42 hours from start of MTX

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received methotrexate during induction cycle 4 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 34 80 34
    Median (Full Range) [µmol/L]
    0.64
    0.64
    0.55
    44. Secondary Outcome
    Title Cyclophosphamide Clearance in Induction Chemotherapy
    Description Cyclophosphamide plasma concentration-time data are collected on day 9 in one cycle of induction chemotherapy. Individual estimates of cyclophosphamide clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during a cycle of induction therapy and had samples collected for PK analysis..
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 91 42
    Median (Full Range) [L/h/m^2]
    2.40
    2.23
    2.25
    45. Secondary Outcome
    Title Cyclophosphamide Clearance in Consolidation Chemotherapy Cycle 1
    Description Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of cyclophosphamide clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 20 4 28
    Median (Full Range) [L/h/m^2]
    2.39
    2.08
    2.43
    46. Secondary Outcome
    Title Cyclophosphamide Clearance in Consolidation Chemotherapy Cycle 2
    Description Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of cyclophosphamide clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 18 3 29
    Median (Full Range) [L/h/m^2]
    2.48
    2.55
    2.37
    47. Secondary Outcome
    Title Cyclophosphamide Apparent Oral Clearance in Maintenance Chemotherapy Cycle A1
    Description Cyclophosphamide plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of cyclophosphamide apparent oral clearance are obtained using post hoc analysis.
    Time Frame Pre-dose, 0.5, 1.75, 3 and 6 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during maintenance cycle A1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 26 42 14
    Median (Full Range) [L/h/m^2]
    2.95
    2.83
    2.74
    48. Secondary Outcome
    Title Cyclophosphamide AUC0-24h in Induction Chemotherapy
    Description Cyclophosphamide plasma concentration-time data are collected on day 9 in one cycle of induction chemotherapy. Individual estimates of cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during a cycle of induction therapy and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 91 42
    Median (Full Range) [µmol·h/L]
    2070
    2150
    2105
    49. Secondary Outcome
    Title Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 1
    Description Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 20 4 28
    Median (Full Range) [µmol·h/L]
    1968
    1504
    868
    50. Secondary Outcome
    Title Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 2
    Description Cyclophosphamide plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 18 3 29
    Median (Full Range) [µmol·h/L]
    1966
    799
    899
    51. Secondary Outcome
    Title Cyclophosphamide AUC0-24h in Maintenance Chemotherapy Cycle A1
    Description Cyclophosphamide plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of cyclophosphamide AUC0-24h are obtained using post hoc analysis.
    Time Frame Pre-dose, 0.5, 1.75, 3, 6, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during maintenance cycle A1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 26 42 14
    Median (Full Range) [µmol·h/L]
    39.9
    38.7
    42.2
    52. Secondary Outcome
    Title 4-OH Cyclophosphamide AUC0-24h in Induction Chemotherapy
    Description 4-OH cyclophosphamide plasma concentration-time data are collected on day 9 in one cycle of induction chemotherapy. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during a cycle of induction therapy and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 91 42
    Median (Full Range) [µmol·h/L]
    116.4
    111.3
    109.1
    53. Secondary Outcome
    Title 4-OH Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 1
    Description 4-OH cyclophosphamide plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 20 4 28
    Median (Full Range) [µmol·h/L]
    96.8
    48.7
    39.8
    54. Secondary Outcome
    Title 4-OH Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 2
    Description 4-OH cyclophosphamide plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 18 3 29
    Median (Full Range) [µmol·h/L]
    95.9
    49.5
    43.5
    55. Secondary Outcome
    Title 4-OH Cyclophosphamide AUC0-24h in Maintenance Chemotherapy Cycle A1
    Description 4-OH cyclophosphamide plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of 4-OH cyclophosphamide AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-dose, 0.5, 1.75, 3, 6, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during maintenance cycle A1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 26 42 14
    Median (Full Range) [µmol·h/L]
    1.98
    1.96
    1.82
    56. Secondary Outcome
    Title CEPM AUC0-24h in Induction Chemotherapy
    Description Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected on day 9 in one induction cycle. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during a cycle of induction therapy and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 38 91 42
    Median (Full Range) [µmol·h/L]
    140.2
    137.8
    135.3
    57. Secondary Outcome
    Title CEPM AUC0-24h in Consolidation Chemotherapy Cycle 1
    Description Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected in consolidation cycle 1. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 20 4 28
    Median (Full Range) [µmol·h/L]
    128.9
    62.2
    51.8
    58. Secondary Outcome
    Title CEPM AUC0-24h in Consolidation Chemotherapy Cycle 2
    Description Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected in consolidation cycle 2. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, end of infusion, 3, 6, and 24 hours from end of cyclophosphamide infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during consolidation cycle 2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 18 3 29
    Median (Full Range) [µmol·h/L]
    132.7
    46.8
    44.0
    59. Secondary Outcome
    Title CEPM AUC0-24h in Maintenance Chemotherapy Cycle A1
    Description Carboxyethylphosphoramide mustard (CEPM) plasma concentration-time data are collected on day 1 of maintenance cycle A1. Individual estimates of CEPM AUC0-24h (area under concentration curve from time 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-dose, 0.5, 1.75, 3, 6, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received cyclophosphamide during maintenance cycle A1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 26 42 14
    Median (Full Range) [µmol·h/L]
    1.59
    1.65
    1.41
    60. Secondary Outcome
    Title Participants With Empirical Dosage Achieving Target System Exposure of Intravenous Topotecan
    Description Number of participants who successfully achieve target systemic exposure of intravenous topotecan after an empiric dosage during consolidation phase of therapy are reported.
    Time Frame Pre-infusion, 5 min., 1, and 3 hours from end of infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received topotecan with empirical dosage during consolidation therapy were included in this analysis. Per protocol, low-risk and intermediate-risk patients did not receive topotecan during consolidation unless they experienced disease progression during induction. One such intermediate-risk patient was included.
    Arm/Group Title Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 1 27
    Number [participants]
    0
    0%
    8
    5.1%
    61. Secondary Outcome
    Title Participants With PK-guided Dosage Adjustment Achieving Target System Exposure of Intravenous Topotecan
    Description Number of participants who successfully achieve target systemic exposure of intravenous topotecan after a pharmacokinetic-guided dosage adjustment during consolidation phase of therapy are reported.
    Time Frame Pre-infusion, 5 min., 1, and 3 hours from end of infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received topotecan with PK-guided dosage adjustment during consolidation therapy were included in this analysis. Per protocol, low-risk and intermediate-risk patients did not receive topotecan during consolidation unless they experienced disease progression during induction. One such intermediate-risk patient was included.
    Arm/Group Title Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 1 27
    Number [participants]
    1
    1.8%
    20
    12.8%
    62. Secondary Outcome
    Title Topotecan Clearance in Consolidation Chemotherapy
    Description Topotecan plasma concentration-time data are collected on day 1 of consolidation cycle 1 after a single IV dose. Individual estimates of topotecan clearance are obtained using post hoc analysis.
    Time Frame Pre-infusion, 5 min., 1, and 3 hours from end of infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received intravenous topotecan during consolidation cycle 1 and had PK samples collected were included in this analysis. Per protocol, low- and intermediate-risk patients did not receive topotecan during consolidation unless they experienced disease progression during induction. One such intermediate-risk patient was included.
    Arm/Group Title Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 1 27
    Median (Full Range) [L/h/m^2]
    30.3
    26.40
    63. Secondary Outcome
    Title Topotecan Apparent Oral Clearance in Maintenance Chemotherapy
    Description Topotecan plasma concentration-time data are collected on day 1 of maintenance cycle A1 after a single oral dose. Individual estimates of topotecan apparent oral clearance are obtained using post hoc analysis.
    Time Frame Pre-dose, 0.25, 1.5 and 6 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received oral topotecan during maintenance therapy cycle A1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 18 33 10
    Median (Full Range) [L/h]
    41.4
    41.0
    44.6
    64. Secondary Outcome
    Title Topotecan AUC0-24h in Consolidation Chemotherapy
    Description Topotecan plasma concentration-time data are collected on day 1 of consolidation cycle 1 after a single IV dose. Individual estimates of topotecan AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.
    Time Frame Pre-infusion, 5 min., 1, 3, and 24 hours from end of infusion

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received intravenous topotecan during consolidation cycle 1 and had PK samples collected were included in this analysis. Per protocol, low- and intermediate-risk patients did not receive topotecan during consolidation unless they experienced disease progression during induction. One such intermediate-risk patient was included.
    Arm/Group Title Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 1 27
    Median (Full Range) [µg·h/L]
    117
    116
    65. Secondary Outcome
    Title Topotecan AUC0-24h in Maintenance Chemotherapy
    Description Topotecan plasma concentration-time data are collected on day 1 of maintenance cycle A1 after a single oral dose. Individual estimates of topotecan AUC0-24h (area under concentration curve from time 0 to 24 hours post- dose) are obtained using post hoc analysis.
    Time Frame Pre-dose, 0.25, 1.5, 6, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received oral topotecan during maintenance therapy cycle A1 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 18 33 10
    Median (Full Range) [µg·h/L]
    10.90
    11.60
    10.33
    66. Secondary Outcome
    Title Erlotinib Apparent Oral Clearance
    Description Erlotinib plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of erlotinib apparent oral clearance are obtained using post hoc analysis.
    Time Frame Pre-dose, 1, 2, 4, 8, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received oral erlotinib during maintenance therapy cycle B2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 10 11 3
    Median (Full Range) [L/h/m^2]
    6.53
    7.79
    8.40
    67. Secondary Outcome
    Title Erlotinib Apparent Volume of Central Compartment
    Description Erlotinib plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of erlotinib apparent volume of central compartment are obtained using post hoc analysis.
    Time Frame Pre-dose, 1, 2, 4, 8, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received oral erlotinib during maintenance therapy cycle B2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 10 11 3
    Median (Full Range) [L/m^2]
    72.9
    61.7
    104.8
    68. Secondary Outcome
    Title Erlotinib AUC0-24h
    Description Erlotinib plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of erlotinib AUC0-24h (area under concentration curve from 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-dose, 1, 2, 4, 8, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received oral erlotinib during maintenance therapy cycle B2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 10 11 3
    Median (Full Range) [µmol·h/L]
    31.0
    23.5
    22.0
    69. Secondary Outcome
    Title OSI-420 AUC0-24h
    Description Erlotinib metabolite OSI-420 plasma concentration-time data are collected on day 1 of maintenance cycle B2. Individual estimates of OSI-420 AUC0-24h (area under concentration curve from 0 to 24 hours post-dose) are obtained using post hoc analysis.
    Time Frame Pre-dose, 1, 2, 4, 8, and 24 hours post-dose

    Outcome Measure Data

    Analysis Population Description
    Eligible patients who received oral erlotinib during maintenance therapy cycle B2 and had samples collected for PK analysis.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic histology, or high grade glioma Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a gross total resection (GTR), other histologic diagnoses with no metastatic disease Patients with central nervous system (CNS) metastatic disease
    Measure Participants 10 11 3
    Median (Full Range) [µmol·h/L]
    2.17
    1.81
    1.62
    70. Secondary Outcome
    Title Rate of Local Disease Progression
    Description Local failure was defined as the interval from end of RT to date of local failure (or combined local + distant failure). Competing events were distant failure or second malignancy. Patients without an event were censored at date of last contact. The 1-year cumulative incidence was estimated and reported with a 95% confidence interval.
    Time Frame 1 year after completion of radiation therapy for last patient

    Outcome Measure Data

    Analysis Population Description
    Eligible intermediate-risk patients who received focal radiation were included in this analysis. Of the 156 intermediate risk patients, 121 started radiation.
    Arm/Group Title Intermediate-risk Patients Who Received Focal Radiation
    Arm/Group Description Patients with no evidence of metastatic disease (M0) medulloblastoma or nodular desmoplastic histology with less than a surgical gross total resection (GTR), other histologic diagnoses with no metastatic disease who received induction chemotherapy and intermediate-risk therapy that included focal radiation.
    Measure Participants 121
    Number (95% Confidence Interval) [Percentage of participants]
    13.2
    23.2%
    71. Secondary Outcome
    Title Rate of Distant Disease Progression
    Description Distant failure was defined as the interval from end of RT to date of distant failure (or combined local + distant failure). Competing events were local failure or second malignancy. Patients without an event were censored at date of last contact. The 1-year cumulative incidence was estimated and reported with a 95% confidence interval.
    Time Frame 1 year after completion of radiation therapy for last patient

    Outcome Measure Data

    Analysis Population Description
    Eligible intermediate-risk patients who received focal radiation were included in this analysis. Of the 156 intermediate risk patients, 121 started radiation.
    Arm/Group Title Intermediate-risk Patients Who Received Focal Radiation
    Arm/Group Description Patients with M0 medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease who received induction chemotherapy and intermediate-risk therapy that included focal radiation.
    Measure Participants 121
    Number (95% Confidence Interval) [percentage of participants]
    25.6
    44.9%

    Adverse Events

    Time Frame All adverse events grades 3, 4, and 5 which occur during treatment and for 30 days after the end of treatment, and events which occur later than 30 days after the end of treatment which are felt to be at least possibly related to treatment, to the time of off study, were collected with some exceptions noted in the additional description section.
    Adverse Event Reporting Description Per protocol, some grade 3 and 4 hematologic toxicities, grade 3 elevation in serum ALT or AST, grade 3 and 4 electrolyte abnormalities, and total parenteral nutrition or intravenous fluids administered to prevent significant weight loss/malnutrition were not collected, especially if they occurred from the beginning of induction chemotherapy. The one patient in the high-risk group who did not start therapy due to early disease progression was excluded from adverse event results.
    Arm/Group Title Low-Risk Group Intermediate-Risk Group High-Risk Group
    Arm/Group Description Patients with gross total resection (GTR)/no evidence of metastatic disease (M0) medulloblastoma, nodular desmoplastic or high grade glioma histology will receive induction chemotherapy and low-risk therapy. Patients with M0 medulloblastoma or nodular desmoplastic histology with less than a GTR, other histologic diagnoses with no metastatic disease, will receive induction chemotherapy and intermediate-risk therapy. Patients with central nervous system (CNS) metastatic disease will receive induction chemotherapy and high-risk therapy.
    All Cause Mortality
    Low-Risk Group Intermediate-Risk Group High-Risk Group
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 10/57 (17.5%) 58/156 (37.2%) 49/77 (63.6%)
    Serious Adverse Events
    Low-Risk Group Intermediate-Risk Group High-Risk Group
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 3/57 (5.3%) 7/156 (4.5%) 5/76 (6.6%)
    Blood and lymphatic system disorders
    Hemoglobin 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Neutrophils/granulocytes (ANC/AGC) 0/57 (0%) 0 0/156 (0%) 0 1/76 (1.3%) 1
    Platelets 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Cardiac disorders
    Hypotension 0/57 (0%) 0 2/156 (1.3%) 2 0/76 (0%) 0
    Ear and labyrinth disorders
    Hearing: patients with/without baseline audiogram and enrolled in a monitoring program 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Gastrointestinal disorders
    Anorexia 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Colitis 0/57 (0%) 0 0/156 (0%) 0 1/76 (1.3%) 1
    Dehydration 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Diarrhea 0/57 (0%) 0 0/156 (0%) 0 1/76 (1.3%) 1
    Mucositis/stomatitis (clinical exam), Oral cavity 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Perforation, GI, Duodenum 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Vomiting 1/57 (1.8%) 1 2/156 (1.3%) 2 1/76 (1.3%) 1
    General disorders
    Fatigue (asthenia, lethargy, malaise) 1/57 (1.8%) 1 0/156 (0%) 0 1/76 (1.3%) 1
    Fever (in the absence of neutropenia, where neutropenia is defined as ANC <1.0 x 10e9/L) 1/57 (1.8%) 1 3/156 (1.9%) 3 0/76 (0%) 0
    Weight loss 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Infections and infestations
    Infection with Grade 3 or 4 neutrophils (ANC <1.0 x 10e9/L), Pharynx 0/57 (0%) 0 0/156 (0%) 0 1/76 (1.3%) 1
    Infection, Upper airway NOS 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Infection - Other 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Infection with normal ANC or Grade 1 or 2 neutrophils, Blood 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Infection with normal ANC or Grade 1 or 2 neutrophils, Catheter-related 1/57 (1.8%) 1 1/156 (0.6%) 1 1/76 (1.3%) 1
    Infection with normal ANC or Grade 1 or 2 neutrophils, Eye NOS 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Infection with normal ANC or Grade 1 or 2 neutrophils, Wound 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Metabolism and nutrition disorders
    Acidosis (metabolic or respiratory) 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Musculoskeletal and connective tissue disorders
    Fracture 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Nervous system disorders
    Hydrocephalus 1/57 (1.8%) 1 0/156 (0%) 0 1/76 (1.3%) 1
    Irritability (children <3 years of age) 1/57 (1.8%) 1 0/156 (0%) 0 0/76 (0%) 0
    Neurology - Other 1/57 (1.8%) 1 0/156 (0%) 0 0/76 (0%) 0
    Neuropathy: motor 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Seizure 1/57 (1.8%) 1 1/156 (0.6%) 1 1/76 (1.3%) 1
    Somnolence/depressed level of consciousness 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Speech impairment (e.g., dysphasia or aphasia) 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Respiratory, thoracic and mediastinal disorders
    Dyspnea (shortness of breath) 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Hypoxia 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Pneumonitis/pulmonary infiltrates 0/57 (0%) 0 1/156 (0.6%) 1 0/76 (0%) 0
    Pulmonary/Upper Respiratory - Other 0/57 (0%) 0 0/156 (0%) 0 2/76 (2.6%) 2
    Vascular disorders
    Hemorrhage/Bleeding - Other 1/57 (1.8%) 1 0/156 (0%) 0 0/76 (0%) 0
    Other (Not Including Serious) Adverse Events
    Low-Risk Group Intermediate-Risk Group High-Risk Group
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 55/57 (96.5%) 145/156 (92.9%) 70/76 (92.1%)
    Blood and lymphatic system disorders
    Febrile neutropenia 36/57 (63.2%) 83 92/156 (59%) 169 48/76 (63.2%) 120
    Neutrophils/granulocytes (ANC/AGC) 32/57 (56.1%) 60 57/156 (36.5%) 124 10/76 (13.2%) 20
    Leukocytes (total WBC) 24/57 (42.1%) 38 45/156 (28.8%) 83 6/76 (7.9%) 11
    Hemoglobin 13/57 (22.8%) 15 9/156 (5.8%) 20 3/76 (3.9%) 3
    Platelets 7/57 (12.3%) 19 8/156 (5.1%) 9 4/76 (5.3%) 4
    Lymphopenia 2/57 (3.5%) 6 8/156 (5.1%) 8 0/76 (0%) 0
    Ear and labyrinth disorders
    Hearing: patients with/without baseline audiogram and enrolled in a monitoring program 3/57 (5.3%) 3 1/156 (0.6%) 1 1/76 (1.3%) 1
    Gastrointestinal disorders
    Vomiting 10/57 (17.5%) 11 25/156 (16%) 32 22/76 (28.9%) 29
    Anorexia 6/57 (10.5%) 7 15/156 (9.6%) 16 12/76 (15.8%) 13
    Diarrhea 6/57 (10.5%) 7 13/156 (8.3%) 14 8/76 (10.5%) 11
    Mucositis/stomatitis (functional/symptomatic), Oral cavity 4/57 (7%) 4 7/156 (4.5%) 8 7/76 (9.2%) 7
    Nausea 1/57 (1.8%) 1 6/156 (3.8%) 6 6/76 (7.9%) 6
    Mucositis/stomatitis (clinical exam), Oral cavity 1/57 (1.8%) 1 11/156 (7.1%) 12 4/76 (5.3%) 5
    Dehydration 3/57 (5.3%) 4 8/156 (5.1%) 9 3/76 (3.9%) 3
    General disorders
    Weight loss 1/57 (1.8%) 1 0/156 (0%) 0 6/76 (7.9%) 6
    Fever (in the absence of neutropenia, where neutropenia is defined as ANC <1.0 x 10e9/L) 0/57 (0%) 0 8/156 (5.1%) 8 3/76 (3.9%) 3
    Infections and infestations
    Infection, Catheter-related 11/57 (19.3%) 14 17/156 (10.9%) 19 8/76 (10.5%) 9
    Infection with normal ANC or Grade 1 or 2 neutrophils, Catheter-related 8/57 (14%) 8 19/156 (12.2%) 26 5/76 (6.6%) 7
    Colitis, infectious (e.g., Clostridium difficile) 3/57 (5.3%) 3 8/156 (5.1%) 12 7/76 (9.2%) 10
    Infection, Skin (cellulitis) 0/57 (0%) 0 7/156 (4.5%) 9 7/76 (9.2%) 7
    Infection, Blood 5/57 (8.8%) 5 5/156 (3.2%) 6 5/76 (6.6%) 5
    Infection - Other 4/57 (7%) 4 8/156 (5.1%) 11 4/76 (5.3%) 5
    Infection with normal ANC or Grade 1 or 2 neutrophils, Blood 2/57 (3.5%) 3 9/156 (5.8%) 9 4/76 (5.3%) 4
    Infection, Upper airway NOS 1/57 (1.8%) 1 4/156 (2.6%) 4 4/76 (5.3%) 4
    Infection, Urinary tract NOS 1/57 (1.8%) 1 2/156 (1.3%) 3 4/76 (5.3%) 4
    Metabolism and nutrition disorders
    ALT, SGPT (serum glutamic pyruvic transaminase) 1/57 (1.8%) 1 8/156 (5.1%) 9 3/76 (3.9%) 3
    Nervous system disorders
    Seizure 1/57 (1.8%) 1 4/156 (2.6%) 5 7/76 (9.2%) 7
    Hydrocephalus 2/57 (3.5%) 2 8/156 (5.1%) 9 5/76 (6.6%) 5
    Respiratory, thoracic and mediastinal disorders
    Hypoxia 1/57 (1.8%) 1 8/156 (5.1%) 10 9/76 (11.8%) 13
    Skin and subcutaneous tissue disorders
    Rash/desquamation 6/57 (10.5%) 7 8/156 (5.1%) 8 2/76 (2.6%) 2

    Limitations/Caveats

    Results for the some of the secondary objectives are not available yet. These results will be posted as they become available.

    More Information

    Certain Agreements

    All Principal Investigators ARE employed by the organization sponsoring the study.

    There is NOT an agreement between Principal Investigators and the Sponsor (or its agents) that restricts the PI's rights to discuss or publish trial results after the trial is completed.

    Results Point of Contact

    Name/Title Amar Gajjar, MD
    Organization St. Jude Children's Research Hospital
    Phone 866-278-5833
    Email amar.gajjar@stjude.org
    Responsible Party:
    St. Jude Children's Research Hospital
    ClinicalTrials.gov Identifier:
    NCT00602667
    Other Study ID Numbers:
    • SJYC07
    • R01CA154619
    • NCI-2011-01193
    First Posted:
    Jan 28, 2008
    Last Update Posted:
    Dec 6, 2021
    Last Verified:
    Dec 1, 2021