Official Title

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

    Phase 2
  • Study Type

    Interventional
  • Study Participants

    293
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.
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 Started
Dec 17
2007
Primary Completion
Sep 27
2017
Study Completion
Apr 30
2026
Anticipated
Results Posted
Jan 30
2019
Last Update
May 10
2023

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)

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: Vepesid(R), VP-16 (etoposide), Velban(R) (vinblastine), Cytoxan(R) (cyclophosphamide), Hycamptin(R) (topotecan), Tarceva(TM) (erlotinib)

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)

Low-Risk Patients Experimental

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.

High-Risk Patients Experimental

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

Intermediate-Risk Therapy Experimental

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.

Criteria

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

Summary

Low-Risk Group

Intermediate-Risk Group

High-Risk Group

All Events

Event Type Organ System Event Term Low-Risk Group Intermediate-Risk Group High-Risk Group

Percent Probability of Progression-free Survival (PFS) for Medulloblastoma Patients

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.

Low-Risk Group

73.9
Percent Probability
95% Confidence Interval: 56.5 to 91.3

Intermediate-Risk Group

46.9
Percent Probability
95% Confidence Interval: 30.2 to 63.6

High-Risk Group

30.8
Percent Probability
95% Confidence Interval: 14.1 to 47.5

Percent Probability of Progression-free Survival (PFS) for Medulloblastoma Patients by DNA Methylation Subgroup

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.

Low-risk SHH Patients

73.9
Percent Probability
95% Confidence Interval: 56.5 to 91.3

Intermediate-risk SHH Patients

50.0
Percent Probability
95% Confidence Interval: 19.0 to 81.0

High-risk SHH Patients

54.5
Percent Probability
95% Confidence Interval: 27.3 to 81.7

Low-risk Group 3 Patients

Intermediate-risk Group 3 Patients

30.8
Percent Probability
95% Confidence Interval: 8.5 to 53.1

High-risk Group 3 Patients

9.1
Percent Probability
95% Confidence Interval: 0.0 to 21.1

Low-risk Group 4 Patients

Intermediate-risk Group 4 Patients

62.5
Percent Probability
95% Confidence Interval: 31.9 to 93.1

High-risk Group 4 Patients

50.0
Percent Probability
95% Confidence Interval: 1.0 to 99.0

Percent Probability of Event-free Survival (EFS) for Medulloblastoma Patients

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.

Low-Risk Group

73.9
Percent Probability
95% Confidence Interval: 56.5 to 91.3

Intermediate-Risk Group

46.9
Percent Probability
95% Confidence Interval: 30.2 to 63.6

High-Risk Group

30.8
Percent Probability
95% Confidence Interval: 14.1 to 47.5

Number of Participants With Chromosomal Abnormalities

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

Low-Risk Group

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

Intermediate-Risk Group

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

High-Risk Group

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

Numbers of Patients With Gene Alterations

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.

Low-Risk Group

BCOR alteration

BRCA2 alteration

GLI2 alteration

KMT2D alteration

MYCN alteration

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

Intermediate-Risk Group

BCOR alteration

BRCA2 alteration

GLI2 alteration

KMT2D alteration

MYCN alteration

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

High-Risk Group

BCOR alteration

BRCA2 alteration

GLI2 alteration

KMT2D alteration

MYCN alteration

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

Numbers of Patients With Molecular Abnormalities by Tumor Type

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.

Low-risk SHH Patients

BCOR alteration

BRCA2 alteration

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

GLI2 alteration

KMT2D alteration

MYCN amplification

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

Intermediate-risk SHH Patients

BCOR alteration

BRCA2 alteration

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

GLI2 alteration

KMT2D alteration

MYCN amplification

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

High-risk SHH Patients

BCOR alteration

BRCA2 alteration

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

GLI2 alteration

KMT2D alteration

MYCN amplification

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

Low-risk Group 3 Patients

Intermediate-risk Group 3 Patients

BCOR alteration

BRCA2 alteration

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

GLI2 alteration

KMT2D alteration

MYCN amplification

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

High-risk Group 3 Patients

BCOR alteration

BRCA2 alteration

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

GLI2 alteration

KMT2D alteration

MYCN amplification

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

Low-risk Group 4 Patients

Intermediate-risk Group 4 Patients

BCOR alteration

BRCA2 alteration

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

GLI2 alteration

KMT2D alteration

MYCN amplification

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

High-risk Group 4 Patients

BCOR alteration

BRCA2 alteration

chr10p gain/amplification

chr10p loss/deletion

chr10q gain/amplification

chr10q loss/deletion

chr20p gain/amplification

chr20p loss/deletion

chr20q gain/amplification

chr20q loss/deletion

chr2p gain/amplification

chr2p loss/deletion

chr2q gain/amplification

chr2q loss/deletion

chr6p gain/amplification

chr6p loss/deletion

chr6q gain/amplification

chr6q loss/deletion

chr8p gain/amplification

chr8p loss/deletion

chr8q gain/amplification

chr8q loss/deletion

chr9p gain/amplification

chr9p loss/deletion

chr9q gain/amplification

chr9q loss/deletion

GLI2 alteration

KMT2D alteration

MYCN amplification

PTCH1 alteration

PTEN alteration

SMARCA4 alteration

SMO alteration

SUFU alteration

TP53 alteration

Number of Successful Collections for Frozen and Fixed Tumor Samples

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

Low-Risk Group

Number with fixed tumor tissue

Number with frozen tumor tissue

Intermediate-Risk Group

Number with fixed tumor tissue

Number with frozen tumor tissue

High-Risk Group

Number with fixed tumor tissue

Number with frozen tumor tissue

Event-free Survival (EFS) Compared to Historical Controls

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.

SJYC07 Low-risk Medulloblastoma Patients

73.9
Percent probability
95% Confidence Interval: 56.5 to 91.3

SJYC07 Intermediate-risk Medulloblastoma Patients

46.9
Percent probability
95% Confidence Interval: 30.2 to 63.6

SJYC07 High-risk Medulloblastoma Patients

30.8
Percent probability
95% Confidence Interval: 14.1 to 47.5

Overall Survival (OS) Compared to Historical Controls

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.

SJYC07 Low-risk Medulloblastoma Patients

100.0
Percent probability
95% Confidence Interval: 100.0 to 100.0

SJYC07 Intermediate-risk Medulloblastoma Patients

84.4
Percent probability
95% Confidence Interval: 72.1 to 96.7

SJYC07 High-risk Medulloblastoma Patients

61.5
Percent probability
95% Confidence Interval: 43.9 to 79.1

Percentage of Patients With Objective Responses Rate to Induction Chemotherapy

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.

Intermediate-Risk Group

58.3
Percentage of patients
95% Confidence Interval: 50.2 to 66.2

High-Risk Group

21.1
Percentage of patients
95% Confidence Interval: 12.5 to 31.8

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

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.

High-Risk Group

3.8
Percentage of courses delayed
95% Confidence Interval: 1.8 to 6.9

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

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.

High-Risk Group

2.6
Percentage of courses delayed
95% Confidence Interval: 0.5 to 8.5

Percent of Patients With Sustained Objective Responses Rate After Consolidation

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.

High-Risk Group

13.2
percentage of participants
95% Confidence Interval: 4.4 to 28.1

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

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.

Low-Risk Group

96.0
Percentage of scheduled doses received (Mean)
Standard Deviation: 8

Intermediate-Risk Group

91.0
Percentage of scheduled doses received (Mean)
Standard Deviation: 23

High-Risk Group

98.0
Percentage of scheduled doses received (Mean)
Standard Deviation: 4

Change in Neurostructure, Especially White Matter Volume and Integrity

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.

Outcome Measure Data Not Reported

Percent of PET Scans With Loss of Signal Intensity

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.

Intermediate Risk Group

60.0
mean activation value (MAV) (Mean)
Standard Deviation: 27

Concentration of Cerebrospinal Fluid Neurotransmitters

Concentrations of various neurotransmitters in cerebrospinal fluid were measured at 5 timepoints. The median concentration of each neurotransmitter at each time point was calculated and provided with a full range.

Patients With Neurotransmitter Studies

3,4-dihydroxyphenylacetic acid concentration at 12 months off treatment

1.04
ng/ml (Median)
Full Range: 0.51 to 6.55

3,4-dihydroxyphenylacetic acid concentration at 24 months off treatment

1.52
ng/ml (Median)
Full Range: 0.31 to 6.7

3,4-dihydroxyphenylacetic acid concentration at 36 months off treatment

1.0
ng/ml (Median)
Full Range: 0.38 to 3.1

3,4-dihydroxyphenylacetic acid concentration at baseline

2.56
ng/ml (Median)
Full Range: 0.91 to 11.24

3,4-dihydroxyphenylacetic acid concentration at completion of treatment

1.62
ng/ml (Median)
Full Range: 0.82 to 9.91

Dopamine concentration at 12 months off treatment

6.43
ng/ml (Median)
Full Range: 2.2 to 486.46

Dopamine concentration at 24 months off treatment

4.46
ng/ml (Median)
Full Range: 0.04 to 1093.28

Dopamine concentration at 36 months off treatment

4.05
ng/ml (Median)
Full Range: 0.11 to 9.32

Dopamine concentration at baseline

3.16
ng/ml (Median)
Full Range: 0.18 to 4586.18

Dopamine concentration at completion of treatment

3.7
ng/ml (Median)
Full Range: 0.82 to 39.16

Homovanillic acid concentration at 12 months off treatment

68.28
ng/ml (Median)
Full Range: 40.78 to 146.84

Homovanillic acid concentration at 24 months off treatment

88.27
ng/ml (Median)
Full Range: 55.27 to 166.99

Homovanillic acid concentration at 36 months off treatment

79.78
ng/ml (Median)
Full Range: 35.4 to 152.51

Homovanillic acid concentration at baseline

82.44
ng/ml (Median)
Full Range: 34.57 to 565.3

Homovanillic acid concentration at completion of treatment

114.13
ng/ml (Median)
Full Range: 73.43 to 179.21

Hydroxyindoleacetic acid concentration at 12 months off treatment

35.72
ng/ml (Median)
Full Range: 24.58 to 51.6

Hydroxyindoleacetic acid concentration at 24 months off treatment

33.98
ng/ml (Median)
Full Range: 25.69 to 45.63

Hydroxyindoleacetic acid concentration at 36 months off treatment

31.56
ng/ml (Median)
Full Range: 16.09 to 53.04

Hydroxyindoleacetic acid concentration at baseline

52.03
ng/ml (Median)
Full Range: 30.02 to 400.35

Hydroxyindoleacetic acid concentration at completion of treatment

52.72
ng/ml (Median)
Full Range: 27.42 to 110.24

Hydroxytryptamine concentration at 12 months off treatment

2.0
ng/ml (Median)
Full Range: 0.06 to 8.46

Hydroxytryptamine concentration at 24 months off treatment

2.44
ng/ml (Median)
Full Range: 1.01 to 7.7

Hydroxytryptamine concentration at 36 months off treatment

1.62
ng/ml (Median)
Full Range: 0.29 to 8.85

Hydroxytryptamine concentration at baseline

2.38
ng/ml (Median)
Full Range: 0.12 to 13.94

Hydroxytryptamine concentration at completion of treatment

2.01
ng/ml (Median)
Full Range: 0.06 to 15.01

Change in Neurocognitive Performance

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

Outcome Measure Data Not Reported

Number and Type of Genetic Polymorphisms

Types of genetic polymorphisms of neurotransmitters were examined. We studied 3 genetic polymorphisms; these were types of genetic polymorphisms involved in dopamine metabolism. They were as follows: rs6323, rs4680, and rs6280.

Number of Patients With Neurotransmitter Studies

rs4680

rs6280

rs6323

Pharmacogenetic Variation on Central Nervous System Transmitters

Frequencies of genetic polymorphisms were reported.

Patients With Neurotransmitter Studies

Genetic Polymorphisms for catecholamine-O-methyltransferase (COMT) rs4680

Genetic Polymorphisms for dopamine receptor D (DRD3) rs6280

Genetic Polymorphisms for monoamine oxidase A (MAOA) rs6323

Change in Neuropsychological Performance

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

Outcome Measure Data Not Reported

Change in Quantitative Magnetic Resonance (MR) Measures in the Frontal Lobe

Outcome Measure Data Not Reported

Change in Neurocognitive Performance in Attention, Working Memory, and Fluency

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.

Outcome Measure Data Not Reported

Change in Quantitative MR Measures in the Right Frontal-parietal Regions

Outcome Measure Data Not Reported

Change in Neurocognitive Performance in Visual-spatial Reasoning and Processing Speed

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

Outcome Measure Data Not Reported

Number of Participants With Endocrinopathy

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.

Outcome Measure Data Not Reported

Longitudinal Change in Growth Hormone Secretion

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.

Outcome Measure Data Not Reported

Methotrexate Clearance in Induction Cycle 1

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.

Low-Risk Group

5.69
L/h/m^2 (Median)
Full Range: 2.19 to 11.08

Intermediate-Risk Group

6.06
L/h/m^2 (Median)
Full Range: 3.07 to 10.69

High-Risk Group

5.65
L/h/m^2 (Median)
Full Range: 1.71 to 9.45

Methotrexate Clearance in Induction Cycle 2

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.

Low-Risk Group

5.47
L/h/m^2 (Median)
Full Range: 2.43 to 10.56

Intermediate-Risk Group

5.7
L/h/m^2 (Median)
Full Range: 2.71 to 8.98

High-Risk Group

5.7
L/h/m^2 (Median)
Full Range: 2.78 to 8.79

Methotrexate Clearance in Induction Cycle 3

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.

Low-Risk Group

5.68
L/h/m^2 (Median)
Full Range: 1.93 to 9.61

Intermediate-Risk Group

5.78
L/h/m^2 (Median)
Full Range: 2.59 to 9.27

High-Risk Group

5.81
L/h/m^2 (Median)
Full Range: 2.78 to 10.2

Methotrexate Clearance in Induction Cycle 4

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.

Low-Risk Group

5.75
L/h/m^2 (Median)
Full Range: 2.14 to 9.65

Intermediate-Risk Group

5.89
L/h/m^2 (Median)
Full Range: 2.95 to 8.84

High-Risk Group

5.79
L/h/m^2 (Median)
Full Range: 2.36 to 9.16

Methotrexate Volume of Central Compartment in Induction Cycle 1

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.

Low-Risk Group

11.63
L/m^2 (Median)
Full Range: 7.09 to 29.58

Intermediate-Risk Group

13.7
L/m^2 (Median)
Full Range: 7.03 to 28.21

High-Risk Group

13.25
L/m^2 (Median)
Full Range: 5.65 to 20.89

Methotrexate Volume of Central Compartment in Induction Cycle 2

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.

Low-Risk Group

13.77
L/m^2 (Median)
Full Range: 5.88 to 28.19

Intermediate-Risk Group

13.73
L/m^2 (Median)
Full Range: 6.47 to 31.45

High-Risk Group

13.62
L/m^2 (Median)
Full Range: 6.87 to 22.39

Methotrexate Volume of Central Compartment in Induction Cycle 3

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.

Low-Risk Group

12.7
L/m^2 (Median)
Full Range: 6.27 to 28.9

Intermediate-Risk Group

13.55
L/m^2 (Median)
Full Range: 7.31 to 24.48

High-Risk Group

13.87
L/m^2 (Median)
Full Range: 6.84 to 22.88

Methotrexate Volume of Central Compartment in Induction Cycle 4

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.

Low-Risk Group

12.64
L/m^2 (Median)
Full Range: 7.16 to 22.92

Intermediate-Risk Group

13.31
L/m^2 (Median)
Full Range: 7.93 to 21.51

High-Risk Group

13.68
L/m^2 (Median)
Full Range: 6.94 to 21.82

Methotrexate AUC0-66h in Induction Cycle 1

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.

Low-Risk Group

1797.0
µmol·h/L (Median)
Full Range: 993.0 to 5029.0

Intermediate-Risk Group

1813.0
µmol·h/L (Median)
Full Range: 1029.0 to 3584.0

High-Risk Group

1821.0
µmol·h/L (Median)
Full Range: 1053.0 to 3631.0

Methotrexate AUC0-66h in Induction Cycle 2

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.

Low-Risk Group

1900.0
µmol·h/L (Median)
Full Range: 1042.0 to 3267.0

Intermediate-Risk Group

1902.0
µmol·h/L (Median)
Full Range: 1225.0 to 3742.0

High-Risk Group

1879.0
µmol·h/L (Median)
Full Range: 1229.0 to 3271.0

Methotrexate AUC0-66h in Induction Cycle 3

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.

Low-Risk Group

1872.0
µmol·h/L (Median)
Full Range: 1145.0 to 3501.0

Intermediate-Risk Group

1879.0
µmol·h/L (Median)
Full Range: 1187.0 to 3756.0

High-Risk Group

1831.0
µmol·h/L (Median)
Full Range: 979.0 to 3139.0

Methotrexate AUC0-66h in Induction Cycle 4

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.

Low-Risk Group

1804.0
µmol·h/L (Median)
Full Range: 1141.0 to 3504.0

Intermediate-Risk Group

1841.0
µmol·h/L (Median)
Full Range: 1245.0 to 3727.0

High-Risk Group

1886.0
µmol·h/L (Median)
Full Range: 1219.0 to 3491.0

Cyclophosphamide Clearance in Consolidation Chemotherapy Cycle 1

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

Low-Risk Group

2.39
L/h/m^2 (Median)
Full Range: 1.65 to 3.44

Intermediate-Risk Group

2.08
L/h/m^2 (Median)
Full Range: 1.97 to 2.62

High-Risk Group

2.43
L/h/m^2 (Median)
Full Range: 1.29 to 3.21

Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 1

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.

Low-Risk Group

0.49
µmol/L (Median)
Full Range: 0.22 to 3.68

Intermediate-Risk Group

0.57
µmol/L (Median)
Full Range: 0.15 to 4.61

High-Risk Group

0.61
µmol/L (Median)
Full Range: 0.14 to 2.09

Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 2

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.

Low-Risk Group

0.75
µmol/L (Median)
Full Range: 0.19 to 3.09

Intermediate-Risk Group

0.72
µmol/L (Median)
Full Range: 0.26 to 7.61

High-Risk Group

0.69
µmol/L (Median)
Full Range: 0.24 to 1.49

Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 3

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.

Low-Risk Group

0.65
µmol/L (Median)
Full Range: 0.2 to 2.88

Intermediate-Risk Group

0.7
µmol/L (Median)
Full Range: 0.16 to 4.34

High-Risk Group

0.58
µmol/L (Median)
Full Range: 0.24 to 1.96

Methotrexate Concentration at 42 Hours Post-dose in Induction Cycle 4

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.

Low-Risk Group

0.64
µmol/L (Median)
Full Range: 0.26 to 3.12

Intermediate-Risk Group

0.64
µmol/L (Median)
Full Range: 0.22 to 3.22

High-Risk Group

0.55
µmol/L (Median)
Full Range: 0.2 to 1.79

Cyclophosphamide Clearance in Induction Chemotherapy

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.

Low-Risk Group

2.4
L/h/m^2 (Median)
Full Range: 1.16 to 9.15

Intermediate-Risk Group

2.23
L/h/m^2 (Median)
Full Range: 1.23 to 8.95

High-Risk Group

2.25
L/h/m^2 (Median)
Full Range: 0.84 to 3.4

Cyclophosphamide Clearance in Consolidation Chemotherapy Cycle 2

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

Low-Risk Group

2.48
L/h/m^2 (Median)
Full Range: 1.97 to 3.35

Intermediate-Risk Group

2.55
L/h/m^2 (Median)
Full Range: 2.19 to 2.93

High-Risk Group

2.37
L/h/m^2 (Median)
Full Range: 1.61 to 3.16

Cyclophosphamide Apparent Oral Clearance in Maintenance Chemotherapy Cycle A1

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.

Low-Risk Group

2.95
L/h/m^2 (Median)
Full Range: 2.05 to 3.89

Intermediate-Risk Group

2.83
L/h/m^2 (Median)
Full Range: 2.23 to 5.14

High-Risk Group

2.74
L/h/m^2 (Median)
Full Range: 2.23 to 4.02

Cyclophosphamide AUC0-24h in Induction Chemotherapy

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.

Low-Risk Group

2070.0
µmol·h/L (Median)
Full Range: 592.0 to 4158.0

Intermediate-Risk Group

2150.0
µmol·h/L (Median)
Full Range: 615.0 to 3430.0

High-Risk Group

2105.0
µmol·h/L (Median)
Full Range: 1523.0 to 5121.0

Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 1

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.

Low-Risk Group

1968.0
µmol·h/L (Median)
Full Range: 1480.0 to 2869.0

Intermediate-Risk Group

1504.0
µmol·h/L (Median)
Full Range: 617.0 to 2511.0

High-Risk Group

868.0
µmol·h/L (Median)
Full Range: 660.0 to 1498.0

Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 2

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.

Low-Risk Group

1966.0
µmol·h/L (Median)
Full Range: 1437.0 to 2519.0

Intermediate-Risk Group

799.0
µmol·h/L (Median)
Full Range: 740.0 to 2303.0

High-Risk Group

899.0
µmol·h/L (Median)
Full Range: 700.0 to 1240.0

Cyclophosphamide AUC0-24h in Maintenance Chemotherapy Cycle A1

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.

Low-Risk Group

39.9
µmol·h/L (Median)
Full Range: 25.3 to 51.3

Intermediate-Risk Group

38.7
µmol·h/L (Median)
Full Range: 21.2 to 54.9

High-Risk Group

42.2
µmol·h/L (Median)
Full Range: 28.0 to 53.0

4-OH Cyclophosphamide AUC0-24h in Induction Chemotherapy

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.

Low-Risk Group

116.4
µmol·h/L (Median)
Full Range: 74.6 to 200.3

Intermediate-Risk Group

111.3
µmol·h/L (Median)
Full Range: 71.4 to 181.0

High-Risk Group

109.1
µmol·h/L (Median)
Full Range: 65.0 to 175.0

4-OH Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 1

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.

High-Risk Group

39.8
µmol·h/L (Median)
Full Range: 25.5 to 59.8

Low-Risk Group

96.8
µmol·h/L (Median)
Full Range: 63.0 to 124.4

Intermediate-Risk Group

48.7
µmol·h/L (Median)
Full Range: 25.8 to 124.6

4-OH Cyclophosphamide AUC0-24h in Consolidation Chemotherapy Cycle 2

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.

Low-Risk Group

95.9
µmol·h/L (Median)
Full Range: 81.7 to 131.1

Intermediate-Risk Group

49.5
µmol·h/L (Median)
Full Range: 39.5 to 84.5

High-Risk Group

43.5
µmol·h/L (Median)
Full Range: 30.8 to 72.5

4-OH Cyclophosphamide AUC0-24h in Maintenance Chemotherapy Cycle A1

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.

Low-Risk Group

1.98
µmol·h/L (Median)
Full Range: 0.98 to 3.01

Intermediate-Risk Group

1.96
µmol·h/L (Median)
Full Range: 1.12 to 3.08

High-Risk Group

1.82
µmol·h/L (Median)
Full Range: 1.19 to 2.4

CEPM AUC0-24h in Induction Chemotherapy

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.

Low-Risk Group

140.2
µmol·h/L (Median)
Full Range: 50.1 to 453.0

Intermediate-Risk Group

137.8
µmol·h/L (Median)
Full Range: 80.4 to 328.9

High-Risk Group

135.3
µmol·h/L (Median)
Full Range: 57.6 to 591.7

CEPM AUC0-24h in Consolidation Chemotherapy Cycle 1

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.

Low-Risk Group

128.9
µmol·h/L (Median)
Full Range: 90.8 to 272.9

Intermediate-Risk Group

62.2
µmol·h/L (Median)
Full Range: 48.8 to 193.9

High-Risk Group

51.8
µmol·h/L (Median)
Full Range: 18.9 to 97.0

CEPM AUC0-24h in Consolidation Chemotherapy Cycle 2

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.

Low-Risk Group

132.7
µmol·h/L (Median)
Full Range: 89.7 to 244.0

Intermediate-Risk Group

46.8
µmol·h/L (Median)
Full Range: 41.9 to 69.0

High-Risk Group

44.0
µmol·h/L (Median)
Full Range: 21.1 to 73.9

CEPM AUC0-24h in Maintenance Chemotherapy Cycle A1

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.

Low-Risk Group

1.59
µmol·h/L (Median)
Full Range: 0.75 to 3.48

Intermediate-Risk Group

1.65
µmol·h/L (Median)
Full Range: 0.68 to 3.64

High-Risk Group

1.41
µmol·h/L (Median)
Full Range: 0.64 to 3.96

Participants With Empirical Dosage Achieving Target System Exposure of Intravenous Topotecan

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

Intermediate-Risk Group

High-Risk Group

8.0
participants

Participants With PK-guided Dosage Adjustment Achieving Target System Exposure of Intravenous Topotecan

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.

Intermediate-Risk Group

1.0
participants

High-Risk Group

20.0
participants

Topotecan Clearance in Consolidation Chemotherapy

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.

Intermediate-Risk Group

30.3
L/h/m^2 (Median)
Full Range: 30.3 to 30.3

High-Risk Group

26.4
L/h/m^2 (Median)
Full Range: 6.8 to 43.0

Topotecan Apparent Oral Clearance in Maintenance Chemotherapy

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.

Low-Risk Group

41.4
L/h (Median)
Full Range: 12.3 to 54.6

Intermediate-Risk Group

41.0
L/h (Median)
Full Range: 24.4 to 62.7

High-Risk Group

44.6
L/h (Median)
Full Range: 18.1 to 57.0

Topotecan AUC0-24h in Consolidation Chemotherapy

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.

Intermediate-Risk Group

117.0
µg·h/L (Median)
Full Range: 117.0 to 117.0

High-Risk Group

116.0
µg·h/L (Median)
Full Range: 90.0 to 262.0

Topotecan AUC0-24h in Maintenance Chemotherapy

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.

Low-Risk Group

10.9
µg·h/L (Median)
Full Range: 9.04 to 24.36

Intermediate-Risk Group

11.6
µg·h/L (Median)
Full Range: 7.93 to 20.5

High-Risk Group

10.33
µg·h/L (Median)
Full Range: 7.96 to 16.61

Erlotinib Apparent Oral Clearance

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.

High-Risk Group

8.4
L/h/m^2 (Median)
Full Range: 5.96 to 10.46

Low-Risk Group

6.53
L/h/m^2 (Median)
Full Range: 3.13 to 18.55

Intermediate-Risk Group

7.79
L/h/m^2 (Median)
Full Range: 3.87 to 16.84

Erlotinib Apparent Volume of Central Compartment

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.

Low-Risk Group

72.9
L/m^2 (Median)
Full Range: 39.5 to 233.7

Intermediate-Risk Group

61.7
L/m^2 (Median)
Full Range: 41.3 to 119.0

High-Risk Group

104.8
L/m^2 (Median)
Full Range: 47.6 to 109.9

Erlotinib AUC0-24h

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.

Low-Risk Group

31.0
µmol·h/L (Median)
Full Range: 7.2 to 64.8

Intermediate-Risk Group

23.5
µmol·h/L (Median)
Full Range: 12.5 to 48.6

High-Risk Group

22.0
µmol·h/L (Median)
Full Range: 13.8 to 33.0

OSI-420 AUC0-24h

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.

Low-Risk Group

2.17
µmol·h/L (Median)
Full Range: 0.73 to 5.98

Intermediate-Risk Group

1.81
µmol·h/L (Median)
Full Range: 1.17 to 6.97

High-Risk Group

1.62
µmol·h/L (Median)
Full Range: 1.25 to 2.93

Rate of Local Disease Progression

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.

Intermediate-risk Patients Who Received Focal Radiation

13.2
Percentage of participants
95% Confidence Interval: 7.2 to 19.3

Rate of Distant Disease Progression

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.

Intermediate-risk Patients Who Received Focal Radiation

25.6
percentage of participants
95% Confidence Interval: 17.8 to 33.4

Total

290
Participants

Age, Continuous

20.8
months (Mean)
Standard Deviation: 11.9

Age, Continuous

20.1
months (Median)
Full Range: 0.2 to 60.6

Race/Ethnicity, Customized

Race/Ethnicity, Customized

Sex: Female, Male

Overall Study

Low-Risk Group

Intermediate-Risk Group

High-Risk Group

Drop/Withdrawal Reasons

Low-Risk Group

Intermediate-Risk Group

High-Risk Group