18 results on '"Gilbertson, Richard J"'
Search Results
2. Brain Tumors: Challenges and Opportunities to Cure.
- Author
-
Mellinghoff, Ingo K. and Gilbertson, Richard J.
- Published
- 2017
- Full Text
- View/download PDF
3. Phase I Trial of MK-0752 in Children With Refractory CNS Malignancies: A Pediatric Brain Tumor Consortium Study.
- Author
-
Fouladi, Maryam, Stewart, Clinton F., Olson, James, Wagner, Lars M., Onar-Thomas, Arzu, Kocak, Mehmet, Packer, Roger J., Goldman, Stewart, Gururangan, Sridharan, Gajjar, Amar, Demuth, Tim, Kun, Larry E., Boyett, James M., and Gilbertson, Richard J.
- Published
- 2011
- Full Text
- View/download PDF
4. Pediatric phase I and pharmacokinetic study of erlotinib followed by the combination of erlotinib and temozolomide: a Children's Oncology Group Phase I Consortium Study.
- Author
-
Jakacki RI, Hamilton M, Gilbertson RJ, Blaney SM, Tersak J, Krailo MD, Ingle AM, Voss SD, Dancey JE, Adamson PC, Jakacki, Regina I, Hamilton, Marta, Gilbertson, Richard J, Blaney, Susan M, Tersak, Jean, Krailo, Mark D, Ingle, Ashish M, Voss, Stephan D, Dancey, Janet E, and Adamson, Peter C
- Published
- 2008
- Full Text
- View/download PDF
5. Finding the Perfect Partner for Medulloblastoma Prognostication.
- Author
-
Gilbertson, Richard J.
- Published
- 2011
- Full Text
- View/download PDF
6. Outcomes by Clinical and Molecular Features in Children With Medulloblastoma Treated With Risk-Adapted Therapy: Results of an International Phase III Trial (SJMB03).
- Author
-
Gajjar A, Robinson GW, Smith KS, Lin T, Merchant TE, Chintagumpala M, Mahajan A, Su J, Bouffet E, Bartels U, Schechter T, Hassall T, Robertson T, Nicholls W, Gururangan S, Schroeder K, Sullivan M, Wheeler G, Hansford JR, Kellie SJ, McCowage G, Cohn R, Fisher MJ, Krasin MJ, Stewart CF, Broniscer A, Buchhalter I, Tatevossian RG, Orr BA, Neale G, Klimo P Jr, Boop F, Srinivasan A, Pfister SM, Gilbertson RJ, Onar-Thomas A, Ellison DW, and Northcott PA
- Subjects
- Adolescent, Cerebellar Neoplasms genetics, Cerebellar Neoplasms mortality, Cerebellar Neoplasms pathology, Child, Child, Preschool, DNA Mutational Analysis, Epigenome, Epigenomics, Female, High-Throughput Nucleotide Sequencing, Humans, Magnetic Resonance Imaging, Male, Medulloblastoma genetics, Medulloblastoma mortality, Medulloblastoma secondary, Predictive Value of Tests, Progression-Free Survival, Risk Assessment, Risk Factors, Time Factors, Young Adult, Biomarkers, Tumor genetics, Cerebellar Neoplasms therapy, DNA Methylation, Medulloblastoma therapy, Mutation
- Abstract
Purpose: SJMB03 (ClinicalTrials.gov identifier: NCT00085202) was a phase III risk-adapted trial that aimed to determine the frequency and clinical significance of biological variants and genetic alterations in medulloblastoma., Patients and Methods: Patients 3-21 years old were stratified into average-risk and high-risk treatment groups based on metastatic status and extent of resection. Medulloblastomas were molecularly classified into subgroups (Wingless [WNT], Sonic Hedgehog [SHH], group 3, and group 4) and subtypes based on DNA methylation profiles and overlaid with gene mutations from next-generation sequencing. Coprimary study end points were (1) to assess the relationship between ERBB2 protein expression in tumors and progression-free survival (PFS), and (2) to estimate the frequency of mutations associated with WNT and SHH tumors. Clinical and molecular risk factors were evaluated, and the most robust were used to model new risk-classification categories., Results: Three hundred thirty eligible patients with medulloblastoma were enrolled. Five-year PFS was 83.2% (95% CI, 78.4 to 88.2) for average-risk patients (n = 227) and 58.7% (95% CI, 49.8 to 69.1) for high-risk patients (n = 103). No association was found between ERBB2 status and PFS in the overall cohort ( P = .74) or when patients were stratified by clinical risk ( P = .71). Mutations in CTNNB1 (96%), DDX3X (37%), and SMARCA4 (24%) were most common in WNT tumors and PTCH1 (38%), TP53 (21%), and DDX3X (19%) in SHH tumors. Methylome profiling classified 53 WNT (17.4%), 48 SHH (15.7%), 65 group 3 (21.3%), and 139 group 4 (45.6%) tumors. A comprehensive clinicomolecular risk factor analysis identified three low-risk groups (WNT, low-risk SHH, and low-risk combined groups 3 and 4) with excellent (5-year PFS > 90%) and two very high-risk groups (high-risk SHH and high-risk combined groups 3 and 4) with poor survival (5-year PFS < 60%)., Conclusion: These results establish a new risk stratification for future medulloblastoma trials.
- Published
- 2021
- Full Text
- View/download PDF
7. Phase I study of temsirolimus in pediatric patients with recurrent/refractory solid tumors.
- Author
-
Spunt SL, Grupp SA, Vik TA, Santana VM, Greenblatt DJ, Clancy J, Berkenblit A, Krygowski M, Ananthakrishnan R, Boni JP, and Gilbertson RJ
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Adolescent, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Cell Cycle Proteins, Child, Child, Preschool, Drug Administration Schedule, Female, Humans, Infant, Infusions, Intravenous, Male, Maximum Tolerated Dose, Neoplasms enzymology, Neoplasms pathology, Phosphoproteins metabolism, Phosphorylation, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Proto-Oncogene Proteins c-akt metabolism, Ribosomal Protein S6 metabolism, Signal Transduction drug effects, Sirolimus administration & dosage, Sirolimus adverse effects, Sirolimus pharmacokinetics, Sirolimus therapeutic use, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases metabolism, Treatment Outcome, United States, Young Adult, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use, Sirolimus analogs & derivatives
- Abstract
Purpose: To determine dose-limiting toxicities, maximum-tolerated dose (MTD), pharmacokinetics, and pharmacodynamics of weekly intravenous temsirolimus, a mammalian target of rapamycin (mTOR) signaling pathway inhibitor, in pediatric patients with recurrent or refractory solid tumors., Patients and Methods: Cohorts of three to six patients 1 to 21 years of age with recurrent or refractory solid tumors were treated with a 1-hour intravenous infusion of temsirolimus weekly for 3 weeks per course at one of four dose levels: 10, 25, 75, or 150 mg/m(2). During the first two courses, pharmacokinetic and pharmacodynamic evaluations (phosphorylation of S6, AKT, and 4EBP1 in peripheral-blood mononuclear cells) were performed., Results: Dose-limiting toxicity (grade 3 anorexia) occurred in one of 18 evaluable patients at the 150 mg/m(2) level, which was determined to be tolerable, and an MTD was not identified. In 13 patients evaluable for response after two courses of therapy, one had complete response (CR; neuroblastoma) and five had stable disease (SD). Four patients (three SDs + one CR) remained on treatment for more than 4 months. The sum of temsirolimus and sirolimus areas under the concentration-time curve was comparable to values in adults. AKT and 4EBP1 phosphorylation were inhibited at all dose levels, particularly after two courses., Conclusion: Weekly intravenous temsirolimus is well tolerated in children with recurrent solid tumors, demonstrates antitumor activity, has pharmacokinetics similar to those in adults, and inhibits the mTOR signaling pathway in peripheral-blood mononuclear cells. Further studies are needed to define the optimal dose for use in combination with other antineoplastic agents in pediatric patients.
- Published
- 2011
- Full Text
- View/download PDF
8. TP53 mutations in favorable-risk Wnt/Wingless-subtype medulloblastomas.
- Author
-
Lindsey JC, Hill RM, Megahed H, Lusher ME, Schwalbe EC, Cole M, Hogg TL, Gilbertson RJ, Ellison DW, Bailey S, and Clifford SC
- Subjects
- Cerebellar Neoplasms mortality, Cerebellar Neoplasms pathology, Cerebellar Neoplasms therapy, Genetic Predisposition to Disease, Humans, Medulloblastoma mortality, Medulloblastoma pathology, Medulloblastoma therapy, Phenotype, Prognosis, Risk Assessment, Risk Factors, Cerebellar Neoplasms genetics, Medulloblastoma genetics, Mutation, Tumor Suppressor Protein p53 genetics, Wnt Proteins genetics
- Published
- 2011
- Full Text
- View/download PDF
9. Integrative genomic analysis of medulloblastoma identifies a molecular subgroup that drives poor clinical outcome.
- Author
-
Cho YJ, Tsherniak A, Tamayo P, Santagata S, Ligon A, Greulich H, Berhoukim R, Amani V, Goumnerova L, Eberhart CG, Lau CC, Olson JM, Gilbertson RJ, Gajjar A, Delattre O, Kool M, Ligon K, Meyerson M, Mesirov JP, and Pomeroy SL
- Subjects
- Algorithms, Child, Gene Expression Regulation, Neoplastic, Genomics, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Polymorphism, Single Nucleotide, Predictive Value of Tests, Prognosis, RNA, Neoplasm genetics, Cerebellar Neoplasms genetics, Cerebellar Neoplasms pathology, DNA-Binding Proteins genetics, Medulloblastoma genetics, Medulloblastoma pathology, Transcription Factors genetics
- Abstract
Purpose: Medulloblastomas are heterogeneous tumors that collectively represent the most common malignant brain tumor in children. To understand the molecular characteristics underlying their heterogeneity and to identify whether such characteristics represent risk factors for patients with this disease, we performed an integrated genomic analysis of a large series of primary tumors., Patients and Methods: We profiled the mRNA transcriptome of 194 medulloblastomas and performed high-density single nucleotide polymorphism array and miRNA analysis on 115 and 98 of these, respectively. Non-negative matrix factorization-based clustering of mRNA expression data was used to identify molecular subgroups of medulloblastoma; DNA copy number, miRNA profiles, and clinical outcomes were analyzed for each. We additionally validated our findings in three previously published independent medulloblastoma data sets., Results: Identified are six molecular subgroups of medulloblastoma, each with a unique combination of numerical and structural chromosomal aberrations that globally influence mRNA and miRNA expression. We reveal the relative contribution of each subgroup to clinical outcome as a whole and show that a previously unidentified molecular subgroup, characterized genetically by c-MYC copy number gains and transcriptionally by enrichment of photoreceptor pathways and increased miR-183∼96∼182 expression, is associated with significantly lower rates of event-free and overall survivals., Conclusion: Our results detail the complex genomic heterogeneity of medulloblastomas and identify a previously unrecognized molecular subgroup with poor clinical outcome for which more effective therapeutic strategies should be developed.
- Published
- 2011
- Full Text
- View/download PDF
10. Phase I study of vandetanib during and after radiotherapy in children with diffuse intrinsic pontine glioma.
- Author
-
Broniscer A, Baker JN, Tagen M, Onar-Thomas A, Gilbertson RJ, Davidoff AM, Pai Panandiker AS, Leung W, Chin TK, Stewart CF, Kocak M, Rowland C, Merchant TE, Kaste SC, and Gajjar A
- Subjects
- Adolescent, Antineoplastic Agents adverse effects, Brain Stem Neoplasms diagnosis, Chemotherapy, Adjuvant, Child, Child, Preschool, Disease-Free Survival, Dose Fractionation, Radiation, Drug Administration Schedule, Female, Glioma diagnosis, Humans, Kaplan-Meier Estimate, Magnetic Resonance Imaging, Male, Maximum Tolerated Dose, Piperidines adverse effects, Quinazolines adverse effects, Radiotherapy, Adjuvant, Treatment Outcome, Young Adult, Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacokinetics, Brain Stem Neoplasms drug therapy, Brain Stem Neoplasms radiotherapy, Glioma drug therapy, Glioma radiotherapy, Piperidines administration & dosage, Piperidines pharmacokinetics, Quinazolines administration & dosage, Quinazolines pharmacokinetics
- Abstract
Purpose: To evaluate the safety, maximum-tolerated dose, pharmacokinetics, and pharmacodynamics of vandetanib, an oral vascular endothelial growth factor receptor 2 (VEGFR2) and epidermal growth factor receptor inhibitor, administered once daily during and after radiotherapy in children with newly diagnosed diffuse intrinsic pontine glioma., Patients and Methods: Radiotherapy was administered as 1.8-Gy fractions (total cumulative dose of 54 Gy). Vandetanib was administered concurrently with radiotherapy for a maximum of 2 years. Dose-limiting toxicities (DLTs) were evaluated during the first 6 weeks of therapy. Pharmacokinetic studies were obtained for all patients. Plasma angiogenic factors and VEGFR2 phosphorylation in mononuclear cells were analyzed before and during therapy., Results: Twenty-one patients were administered 50 (n = 3), 65 (n = 3), 85 (n = 3), 110 (n = 6), and 145 mg/m(2) (n = 6) of vandetanib. Only one patient developed DLT (grade 3 diarrhea) at dosage level 5. An expanded cohort of patients were treated at dosage levels 4 (n = 10) and 5 (n = 4); two patients developed grade 4 hypertension and posterior reversible encephalopathy syndrome while also receiving high-dose dexamethasone. Despite significant interpatient variability, exposure to vandetanib increased with higher dosage levels. The bivariable analysis of vascular endothelial growth factor (VEGF) before and during therapy showed that patients with higher levels of VEGF before therapy had a longer progression-free survival (PFS; P = .022), whereas patients with increases in VEGF during treatment had a shorter PFS (P = .0015). VEGFR2 phosphorylation was inhibited on day 8 or 29 of therapy compared with baseline (P = .039)., Conclusion: The recommended phase II dose of vandetanib in children is 145 mg/m(2) per day. Close monitoring and management of hypertension is required, particularly for patients receiving corticosteroids.
- Published
- 2010
- Full Text
- View/download PDF
11. Phase I trial of lapatinib in children with refractory CNS malignancies: a Pediatric Brain Tumor Consortium study.
- Author
-
Fouladi M, Stewart CF, Blaney SM, Onar-Thomas A, Schaiquevich P, Packer RJ, Gajjar A, Kun LE, Boyett JM, and Gilbertson RJ
- Subjects
- Administration, Oral, Adolescent, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Central Nervous System Neoplasms chemistry, Child, Child, Preschool, Diarrhea chemically induced, ErbB Receptors analysis, ErbB Receptors antagonists & inhibitors, Exanthema chemically induced, Fatigue chemically induced, Female, Humans, Infant, Lapatinib, Male, Maximum Tolerated Dose, Phosphorylation, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Quinazolines adverse effects, Quinazolines pharmacokinetics, Receptor, ErbB-2 analysis, Receptor, ErbB-2 antagonists & inhibitors, Steroids therapeutic use, Treatment Outcome, United States, Young Adult, Antineoplastic Agents administration & dosage, Central Nervous System Neoplasms drug therapy, Protein Kinase Inhibitors administration & dosage, Quinazolines administration & dosage
- Abstract
Purpose: To estimate the maximum-tolerated dose, dose-limiting toxicities (DLTs), and pharmacokinetic properties of lapatinib, a selective epidermal growth factor receptor (EGFR) and ERBB2 inhibitor, in children with refractory or recurrent CNS malignancies., Patients and Methods: Lapatinib was administered orally twice daily at escalating doses starting at 300 mg/m(2) to patients who were not (stratum I) or were (stratum II) receiving steroids. Pharmacokinetic studies were performed during the first two courses. Expression of the four ERBB receptors and downstream signaling elements in tumor tissue was evaluated by immunohistochemistry., Results: Fifty-nine patients were enrolled (stratum I, n = 32; stratum II, n = 27). Of 29 patients evaluable for toxicity in stratum I, one experienced a DLT (diarrhea) at 520 mg/m(2) twice daily, and all three receiving 1,150 mg/m(2) twice daily experienced DLTs (one each of rash, diarrhea, and fatigue). Two of 21 patients evaluable for toxicity in stratum II experienced DLTs of rash at 900 mg/m(2) twice daily. Lapatinib dosage was related linearly to area under the [concentration-time] curve from start time to 12 hours later (AUC(0-12)) and dose-normalized maximum serum concentration and AUC values for patients in stratum II were both significantly higher (P = .001) than those for patients in stratum I. Frequent, high-level expression of activated (phosphorylated) EGFR and ERBB2 receptors and downstream signal intermediates were observed in tumors, particularly in ependymomas that displayed prolonged stable disease on lapatinib therapy., Conclusion: Lapatinib is well tolerated in children with recurrent CNS malignancies, with rash, diarrhea, and fatigue identified as DLTs. The recommended phase II dose, regardless of steroid use, is 900 mg/m(2) twice daily.
- Published
- 2010
- Full Text
- View/download PDF
12. Pediatric phase I trial and pharmacokinetic study of vorinostat: a Children's Oncology Group phase I consortium report.
- Author
-
Fouladi M, Park JR, Stewart CF, Gilbertson RJ, Schaiquevich P, Sun J, Reid JM, Ames MM, Speights R, Ingle AM, Zwiebel J, Blaney SM, and Adamson PC
- Subjects
- Administration, Oral, Adolescent, Adult, Antineoplastic Agents adverse effects, Child, Child, Preschool, Female, Humans, Hydroxamic Acids adverse effects, Hypokalemia chemically induced, Male, Maximum Tolerated Dose, Neoplasms metabolism, Neutropenia chemically induced, Thrombocytopenia chemically induced, Vorinostat, Antineoplastic Agents administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Hydroxamic Acids administration & dosage, Hydroxamic Acids pharmacokinetics, Neoplasms drug therapy, Tretinoin administration & dosage
- Abstract
Purpose: The purpose of this study was to determine the maximum-tolerated dose (MTD), dose-limiting toxicities (DLT), and pharmacokinetics of vorinostat administered as a single agent and in combination 13-cis retinoic acid (13cRA) in children with refractory solid tumors; to evaluate the tolerability of the solid tumor MTD in children with refractory leukemias; and to characterize the pharmacokinetics of a vorinostat suspension in children., Patients and Methods: Vorinostat was administered orally daily starting at 180 mg/m(2)/d with escalations planned in 30% increments. Pharmacokinetic studies were performed with the initial dose. Acetyl-histone (H3) accumulation was assessed by Western blotting of peripheral blood mononuclear cells (PBMC)., Results: Sixty-four patients were enrolled on this multipart trial. In patients with solid tumors, the MTD was 230 mg/m(2)/d with dose-limiting neutropenia, thrombocytopenia, and hypokalemia at 300 mg/m(2)/d. DLTs observed with the combination of 13cRA and vorinostat included thrombocytopenia, neutropenia, anorexia, and hypertriglyceridemia, resulting in a MTD of vorinostat 180 mg/m(2)/d 4 times per week and 13cRA 80 mg/m(2)/dose twice per day, days 1 through 14 every 28 days. Wide interpatient variability was noted in vorinostat disposition, with area under the concentration-time curves at 230 mg/m(2)/d for the capsule (range, 1,415 to 9,291 ng/mL x hr) and oral suspension (range, 1,186 to 4,780 ng/mL x hr). Significant accumulation of acetylated H3 histone in PBMC was observed after administration of vorinostat, particularly at higher doses. One patient with neuroblastoma experienced a complete response to the combination., Conclusion: In children with recurrent solid tumors, vorinostat is well-tolerated at 230 mg/m(2)/d, with a modest dose reduction being required when combining vorinostat with 13cRA. Drug disposition is similar to that observed in adults.
- Published
- 2010
- Full Text
- View/download PDF
13. Lack of efficacy of bevacizumab plus irinotecan in children with recurrent malignant glioma and diffuse brainstem glioma: a Pediatric Brain Tumor Consortium study.
- Author
-
Gururangan S, Chi SN, Young Poussaint T, Onar-Thomas A, Gilbertson RJ, Vajapeyam S, Friedman HS, Packer RJ, Rood BN, Boyett JM, and Kun LE
- Subjects
- Adolescent, Adult, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized, Bevacizumab, Brain Neoplasms metabolism, Brain Neoplasms pathology, Brain Stem Neoplasms metabolism, Brain Stem Neoplasms pathology, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Child, Diffusion Magnetic Resonance Imaging, Glioma metabolism, Glioma pathology, Humans, Irinotecan, Neoplasm Recurrence, Local metabolism, Neoplasm Recurrence, Local pathology, Phosphorylation, Survival Rate, Treatment Outcome, Vascular Endothelial Growth Factor Receptor-2 metabolism, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Brain Stem Neoplasms drug therapy, Glioma drug therapy, Neoplasm Recurrence, Local drug therapy
- Abstract
Purpose: A phase II study of bevacizumab (BVZ) plus irinotecan (CPT-11) was conducted in children with recurrent malignant glioma (MG) and intrinsic brainstem glioma (BSG)., Patients and Methods: Eligible patients received two doses of BVZ intravenously (10 mg/kg) 2 weeks apart and then BVZ plus CPT-11 every 2 weeks until progressive disease, unacceptable toxicity, or a maximum of 2 years of therapy. Correlative studies included diffusion weighted and T1 dynamic contrast-enhanced permeability imaging, BVZ pharmacokinetics, and estimation of vascular endothelial growth factor receptor 2 (VEGFR-2) phosphorylation in peripheral blood mononuclear cells (PBMC) after single-agent BVZ., Results: Thirty-one evaluable patients received a median of two courses of BVZ plus CPT-11 (range, 1 to 19). No sustained responses were observed in either stratum. Median time to progression for all 34 eligible patients enrolled was 127 days for MG and 71 days for BSG. Progression-free survival rates at 6 months were 41.8% and 9.7% for MG and BSG, respectively. Toxicities related to BVZ included grade 1 to 3 fatigue in seven patients, grade 1 to 2 hypertension in seven patients, grade 1 CNS hemorrhage in four patients, and grade 4 CNS ischemia in two patients. The mean diffusion ratio decreased after two doses of BVZ in patients with MG only. Vascular permeability parameters did not change significantly after therapy in either stratum. Inhibition of VEGFR-2 phosphorylation in PBMC was detected in eight of 11 patients after BVZ exposure., Conclusion: BVZ plus CPT-11 was well-tolerated but had minimal efficacy in children with recurrent malignant glioma and brainstem glioma.
- Published
- 2010
- Full Text
- View/download PDF
14. Phase I study of everolimus in pediatric patients with refractory solid tumors.
- Author
-
Fouladi M, Laningham F, Wu J, O'Shaughnessy MA, Molina K, Broniscer A, Spunt SL, Luckett I, Stewart CF, Houghton PJ, Gilbertson RJ, and Furman WL
- Subjects
- Administration, Oral, Adolescent, Adult, Antineoplastic Agents pharmacokinetics, Child, Child, Preschool, Dose-Response Relationship, Drug, Drug Administration Schedule, Everolimus, Female, Humans, Male, Maximum Tolerated Dose, Neoplasms metabolism, Sirolimus pharmacokinetics, Sirolimus therapeutic use, Survival Rate, Treatment Outcome, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Salvage Therapy, Sirolimus analogs & derivatives
- Abstract
Purpose: To determine the maximum-tolerated dose (MTD), dose-limiting toxicities (DLTs), and pharmacokinetic and pharmacodynamic properties of the mammalian target of rapamycin (mTOR) inhibitor, everolimus, in children with refractory or recurrent solid tumors., Patients and Methods: Everolimus was administered orally at a daily dose of 2.1, 3, 5, or 6.5 mg/m2 in cohorts of three to six patients per dosage level. Pharmacokinetic and pharmacodynamic studies were performed during the first course. The phosphorylation status of various components of the mTOR signal pathway was assessed in peripheral-blood mononuclear cells (PBMCs) isolated from treated patients., Results: There were 26 patients enrolled; 18 were assessable. DLTs included diarrhea (n = 1), mucositis (n = 1), and elevation of ALT (n = 1) at 6.5 mg/m2. At the MTD of 5 mg/m2, the median everolimus clearance was 15.2 L/h/m2, with a plasma everolimus concentration-time area under the curve (AUC) from 0 to infinity of 239.6 ng/mL x h. Significant inhibition of mTOR pathway signaling was observed in PBMCs from patients achieving AUCs 200 ng/mL x h, equivalent to dosages of 3 to 5 mg/m2 of everolimus. No objective tumor responses were observed., Conclusion: Continuous, orally administered everolimus is well tolerated in children with recurrent or refractory solid tumors and demonstrates similar pharmacokinetic properties to those observed in adults. Everolimus significantly inhibits the mTOR signaling pathway in children at the MTD. The recommended phase II dose in children with solid tumors is 5 mg/m2.
- Published
- 2007
- Full Text
- View/download PDF
15. Clinical and molecular characteristics of malignant transformation of low-grade glioma in children.
- Author
-
Broniscer A, Baker SJ, West AN, Fraser MM, Proko E, Kocak M, Dalton J, Zambetti GP, Ellison DW, Kun LE, Gajjar A, Gilbertson RJ, and Fuller CE
- Subjects
- Biopsy, Needle, Brain Neoplasms mortality, Child, Child, Preschool, Cohort Studies, Female, Gene Expression Regulation, Neoplastic, Glioma mortality, Glioma radiotherapy, Humans, Immunohistochemistry, In Situ Hybridization, Male, Neoplasm Staging, Probability, Prognosis, Retrospective Studies, Risk Assessment, Sensitivity and Specificity, Statistics, Nonparametric, Survival Analysis, Treatment Outcome, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Transformation, Neoplastic genetics, Glioma genetics, Glioma pathology, Tumor Suppressor Protein p53 genetics
- Abstract
Purpose: To analyze the clinical and molecular characteristics of malignant transformation (MT) of low-grade glioma (LGG) in children., Patients and Methods: The clinical, radiologic, and histologic characteristics of children treated at our institution who experienced MT of LGG were reviewed. Molecular alterations in these tumors were analyzed by fluorescent in situ hybridization, immunohistochemistry, and TP53 sequencing. Cumulative incidence estimate and risk factors for MT were determined for 65 patients with grade 2 astrocytoma treated at our institution during the study interval., Results: Eleven patients who experienced MT were identified (median age at diagnosis of LGG, 13.3 years). Initial diagnoses were grade 2 astrocytoma (n = 6) and other grade 1/2 gliomas (n = 5). The median latency of MT was 5.1 years. Histologic diagnoses after MT were glioblastoma (n = 7) and other high-grade gliomas (n = 4). The 15-year cumulative incidence estimate of MT among 65 patients with grade 2 astrocytoma was 6.7% +/- 3.9%; no risk factor analyzed, including radiotherapy, was associated with MT. Tissue was available for molecular analysis in all patients, including nine with samples obtained before and after MT. TP53 overexpression was more common after MT. Deletions of RB1 and/or CDKN2A were observed in 71% of LGGs and in 90% of tumors after MT. PTEN pathway abnormalities occurred in 76% of patients. One of five oncogenes analyzed (PDGFRA) was amplified in one patient., Conclusion: The molecular abnormalities that occur during MT of LGG in children are similar to those observed in primary and secondary glioblastoma in adults.
- Published
- 2007
- Full Text
- View/download PDF
16. Genomics identifies medulloblastoma subgroups that are enriched for specific genetic alterations.
- Author
-
Thompson MC, Fuller C, Hogg TL, Dalton J, Finkelstein D, Lau CC, Chintagumpala M, Adesina A, Ashley DM, Kellie SJ, Taylor MD, Curran T, Gajjar A, and Gilbertson RJ
- Subjects
- Adolescent, Adult, Antineoplastic Agents therapeutic use, Cerebellar Neoplasms drug therapy, Cerebellar Neoplasms pathology, Child, Child, Preschool, Cost-Benefit Analysis, DNA Mutational Analysis, Female, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Infant, Infant, Newborn, Male, Medulloblastoma drug therapy, Medulloblastoma pathology, Patient Selection, Reverse Transcriptase Polymerase Chain Reaction, Cerebellar Neoplasms genetics, Gene Expression Profiling economics, Genomics, Medulloblastoma genetics
- Abstract
Purpose: Traditional genetic approaches to identify gene mutations in cancer are expensive and laborious. Nonetheless, if we are to avoid rejecting effective molecular targeted therapies, we must test these drugs in patients whose tumors harbor mutations in the drug target. We hypothesized that gene expression profiling might be a more rapid and cost-effective method of identifying tumors that contain specific genetic abnormalities., Materials and Methods: Gene expression profiles of 46 samples of medulloblastoma were generated using the U133av2 Affymetrix oligonucleotide array and validated using real-time reverse transcriptase polymerase chain reaction (RT-PCR) and immunohistochemistry. Genetic abnormalities were confirmed using fluorescence in situ hybridization (FISH) and direct sequencing., Results: Unsupervised analysis of gene expression profiles partitioned medulloblastomas into five distinct subgroups (subgroups A to E). Gene expression signatures that distinguished these subgroups predicted the presence of key molecular alterations that we subsequently confirmed by gene sequence analysis and FISH. Subgroup-specific abnormalities included mutations in the Wingless (WNT) pathway and deletion of chromosome 6 (subgroup B) and mutations in the Sonic Hedgehog (SHH) pathway (subgroup D). Real-time RT-PCR analysis of gene expression profiles was then used to predict accurately the presence of mutations in the WNT and SHH pathways in a separate group of 31 medulloblastomas., Conclusion: Genome-wide expression profiles can partition large tumor cohorts into subgroups that are enriched for specific genetic alterations. This approach may assist ultimately in the selection of patients for future clinical trials of molecular targeted therapies.
- Published
- 2006
- Full Text
- View/download PDF
17. Atypical teratoid/rhabdoid tumors (ATRT): improved survival in children 3 years of age and older with radiation therapy and high-dose alkylator-based chemotherapy.
- Author
-
Tekautz TM, Fuller CE, Blaney S, Fouladi M, Broniscer A, Merchant TE, Krasin M, Dalton J, Hale G, Kun LE, Wallace D, Gilbertson RJ, and Gajjar A
- Subjects
- Central Nervous System Neoplasms mortality, Central Nervous System Neoplasms pathology, Child, Preschool, Combined Modality Therapy, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Infant, Neoplasms, Germ Cell and Embryonal mortality, Neoplasms, Germ Cell and Embryonal pathology, Prognosis, Rhabdoid Tumor mortality, Rhabdoid Tumor pathology, Survival Rate, Teratoma mortality, Teratoma pathology, Transcription Factors analysis, Central Nervous System Neoplasms therapy, Neoplasms, Germ Cell and Embryonal therapy, Rhabdoid Tumor therapy, Teratoma therapy
- Abstract
Purpose: To describe clinical features, therapeutic approaches, and prognostic factors in pediatric patients with atypical teratoid/rhabdoid tumors (ATRT) treated at St Jude Children's Research Hospital (SJCRH)., Patients and Methods: Primary tumor samples from patients diagnosed with ATRT at SJCRH between July 1984 and June 2003 were identified. Pathology review included histologic, immunohistochemical analysis, and fluorescence in situ hybridization for SMARCB1 (also known as hSNF5/INI1) deletion. Clinical records of patients with pathologic confirmation of ATRT were reviewed., Results: Thirty-seven patients were diagnosed with ATRT at SJCRH during the 19-year study interval. Six patients were excluded from this clinical review based on pathologic or clinical criteria. Of the remaining 31 patients, 22 were younger than 3 years. Posterior fossa primary lesions and metastatic disease at diagnosis were more common in younger patients with ATRT. All patients underwent surgical resection; 30 received subsequent chemotherapy. The majority of patients aged 3 years or older received postoperative craniospinal radiation. Two-year event-free (EFS) and overall survival (OS) of children aged 3 years or older (EFS, 78% + 14%; OS, 89% +/- 11%) were significantly better than those for younger patients (EFS, 11% +/- 6%; OS, 17% +/- 8%); EFS, P = .009 and OS, P = .0001. No other clinical characteristics were predictive of survival. Three of four patients 3 years or older with progressive disease were successfully rescued with ifosfamide, carboplatin, and etoposide therapy., Conclusion: Children presenting with ATRT before the age of 3 years have a dismal prognosis. ATRT presenting in older patients can be cured using a combination of radiation and high-dose alkylating therapy. Older patients with relapsed ATRT can have salvage treatment using ICE chemotherapy.
- Published
- 2005
- Full Text
- View/download PDF
18. Clinical, histopathologic, and molecular markers of prognosis: toward a new disease risk stratification system for medulloblastoma.
- Author
-
Gajjar A, Hernan R, Kocak M, Fuller C, Lee Y, McKinnon PJ, Wallace D, Lau C, Chintagumpala M, Ashley DM, Kellie SJ, Kun L, and Gilbertson RJ
- Subjects
- Adolescent, Australia, Biomarkers, Tumor genetics, Cerebellar Neoplasms genetics, Child, Child, Preschool, Disease-Free Survival, Feasibility Studies, Female, Humans, Infant, Male, Medulloblastoma genetics, Pathology, Clinical organization & administration, Predictive Value of Tests, Prognosis, Proportional Hazards Models, Receptor, ErbB-2 analysis, Risk Assessment, Risk Factors, Specimen Handling, Survival Analysis, United States, Biomarkers, Tumor analysis, Cerebellar Neoplasms pathology, Medulloblastoma pathology
- Abstract
Purpose: To assess the feasibility of performing central molecular analyses of fresh medulloblastomas obtained from multiple institutions and using these data to identify prognostic markers for contemporaneously treated patients., Materials and Methods: Ninety-seven samples of medulloblastoma were collected. Tumor content in samples was judged by frozen section review. Tumor ERBB2 protein and MYCC, MYCN, and TRKC mRNA levels were measured blind to clinical details using Western blotting and real-time polymerase chain reaction, respectively. Histopathologic and clinical review of each case was also performed. All data were subjected to independent statistical analysis., Results: Sample acquisition and analysis times ranged from 3 to 6 days. Eighty-six samples contained sufficient tumor for analysis, including 38 classic, 30 nodular desmoplastic, and 18 large-cell anaplastic (LCA) medulloblastomas. Protein and mRNA were extracted from 81 and 49 tumors, respectively. ERBB2 was detected in 40% (n=32 of 81) of tumors, most frequently in LCA disease (P=.005), and was independently associated with a poor prognosis (P=.031). A combination of clinical characteristics and ERBB2 expression provided a highly accurate means of discriminating disease risk. One hundred percent (n=26) of children with clinical average-risk, ERBB2-negative disease were alive at 5 years, with a median follow-up of 5.6 years, compared with only 54% for children with average-risk, ERBB2-positive tumors (n=13; P=.0001). TRKC, MYCC, and MYCN expression and histopathologic subtype were not associated with prognosis in this study., Conclusion: Central and rapid molecular analysis of frozen medulloblastomas collected from multiple institutions is feasible. ERBB2 expression and clinical risk factors together constitute a highly accurate disease risk stratification tool.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.