Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
2.
Neuro Oncol ; 26(1): 178-190, 2024 01 05.
Article in English | MEDLINE | ID: mdl-37503880

ABSTRACT

BACKGROUND: High-grade gliomas (HGG) in young children pose a challenge due to favorable but unpredictable outcomes. While retrospective studies broadened our understanding of tumor biology, prospective data is lacking. METHODS: A cohort of children with histologically diagnosed HGG from the SJYC07 trial was augmented with nonprotocol patients with HGG treated at St. Jude Children's Research Hospital from November 2007 to December 2020. DNA methylome profiling and whole genome, whole exome, and RNA sequencing were performed. These data were integrated with histopathology to yield an integrated diagnosis. Clinical characteristics and preoperative imaging were analyzed. RESULTS: Fifty-six children (0.0-4.4 years) were identified. Integrated analysis split the cohort into four categories: infant-type hemispheric glioma (IHG), HGG, low-grade glioma (LGG), and other-central nervous system (CNS) tumors. IHG was the most prevalent (n = 22), occurred in the youngest patients (median age = 0.4 years), and commonly harbored receptor tyrosine kinase gene fusions (7 ALK, 2 ROS1, 3 NTRK1/2/3, 4 MET). The 5-year event-free (EFS) and overall survival (OS) for IHG was 53.13% (95%CI: 35.52-79.47) and 90.91% (95%CI: 79.66-100.00) vs. 0.0% and 16.67% (95%CI: 2.78-99.74%) for HGG (p = 0.0043, p = 0.00013). EFS and OS were not different between IHG and LGG (p = 0.95, p = 0.43). Imaging review showed IHGs are associated with circumscribed margins (p = 0.0047), hemispheric location (p = 0.0010), and intratumoral hemorrhage (p = 0.0149). CONCLUSIONS: HGG in young children is heterogeneous and best defined by integrating histopathological and molecular features. Patients with IHG have relatively good outcomes, yet they endure significant deficits, making them good candidates for therapy de-escalation and trials of molecular targeted therapy.


Subject(s)
Brain Neoplasms , Glioma , Child , Infant , Humans , Child, Preschool , Retrospective Studies , Prospective Studies , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Glioma/drug therapy , Glioma/genetics , Glioma/diagnosis , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics
3.
Nat Med ; 30(1): 207-217, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37978284

ABSTRACT

BRAF genomic alterations are the most common oncogenic drivers in pediatric low-grade glioma (pLGG). Arm 1 (n = 77) of the ongoing phase 2 FIREFLY-1 (PNOC026) trial investigated the efficacy of the oral, selective, central nervous system-penetrant, type II RAF inhibitor tovorafenib (420 mg m-2 once weekly; 600 mg maximum) in patients with BRAF-altered, relapsed/refractory pLGG. Arm 2 (n = 60) is an extension cohort, which provided treatment access for patients with RAF-altered pLGG after arm 1 closure. Based on independent review, according to Response Assessment in Neuro-Oncology High-Grade Glioma (RANO-HGG) criteria, the overall response rate (ORR) of 67% met the arm 1 prespecified primary endpoint; median duration of response (DOR) was 16.6 months; and median time to response (TTR) was 3.0 months (secondary endpoints). Other select arm 1 secondary endpoints included ORR, DOR and TTR as assessed by Response Assessment in Pediatric Neuro-Oncology Low-Grade Glioma (RAPNO) criteria and safety (assessed in all treated patients and the primary endpoint for arm 2, n = 137). The ORR according to RAPNO criteria (including minor responses) was 51%; median DOR was 13.8 months; and median TTR was 5.3 months. The most common treatment-related adverse events (TRAEs) were hair color changes (76%), elevated creatine phosphokinase (56%) and anemia (49%). Grade ≥3 TRAEs occurred in 42% of patients. Nine (7%) patients had TRAEs leading to discontinuation of tovorafenib. These data indicate that tovorafenib could be an effective therapy for BRAF-altered, relapsed/refractory pLGG. ClinicalTrials.gov registration: NCT04775485 .


Subject(s)
Fireflies , Glioma , Humans , Child , Animals , Proto-Oncogene Proteins B-raf/genetics , Glioma/drug therapy , Glioma/genetics
4.
Clin Cancer Res ; 29(23): 4770-4783, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37126021

ABSTRACT

PURPOSE: Checkpoint inhibitors have limited efficacy for children with unselected solid and brain tumors. We report the first prospective pediatric trial (NCT02992964) using nivolumab exclusively for refractory nonhematologic cancers harboring tumor mutation burden (TMB) ≥5 mutations/megabase (mut/Mb) and/or mismatch repair deficiency (MMRD). PATIENTS AND METHODS: Twenty patients were screened, and 10 were ultimately included in the response cohort of whom nine had TMB >10 mut/Mb (three initially eligible based on MMRD) and one patient had TMB between 5 and 10 mut/Mb. RESULTS: Delayed immune responses contributed to best overall response of 50%, improving on initial objective responses (20%) and leading to 2-year overall survival (OS) of 50% [95% confidence interval (CI), 27-93]. Four children, including three with refractory malignant gliomas are in complete remission at a median follow-up of 37 months (range, 32.4-60), culminating in 2-year OS of 43% (95% CI, 18.2-100). Biomarker analyses confirmed benefit in children with germline MMRD, microsatellite instability, higher activated and lower regulatory circulating T cells. Stochastic mutation accumulation driven by underlying germline MMRD impacted the tumor microenvironment, contributing to delayed responses. No benefit was observed in the single patient with an MMR-proficient tumor and TMB 7.4 mut/Mb. CONCLUSIONS: Nivolumab resulted in durable responses and prolonged survival for the first time in a pediatric trial of refractory hypermutated cancers including malignant gliomas. Novel biomarkers identified here need to be translated rapidly to clinical care to identify children who can benefit from checkpoint inhibitors, including upfront management of cancer. See related commentary by Mardis, p. 4701.


Subject(s)
Brain Neoplasms , Glioma , Humans , Child , Nivolumab/therapeutic use , Prospective Studies , Mutation , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Biomarkers, Tumor/genetics , DNA Mismatch Repair/genetics , Tumor Microenvironment
5.
J Clin Oncol ; 39(7): 807-821, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33502920

ABSTRACT

PURPOSE: We sought to investigate clinical outcomes of relapsed medulloblastoma and to compare molecular features between patient-matched diagnostic and relapsed tumors. METHODS: Children and infants enrolled on either SJMB03 (NCT00085202) or SJYC07 (NCT00602667) trials who experienced medulloblastoma relapse were analyzed for clinical outcomes, including anatomic and temporal patterns of relapse and postrelapse survival. A largely independent, paired molecular cohort was analyzed by DNA methylation array and next-generation sequencing. RESULTS: A total of 72 of 329 (22%) SJMB03 and 52 of 79 (66%) SJYC07 patients experienced relapse with significant representation of Group 3 and wingless tumors. Although most patients exhibited some distal disease (79%), 38% of patients with sonic hedgehog tumors experienced isolated local relapse. Time to relapse and postrelapse survival varied by molecular subgroup with longer latencies for patients with Group 4 tumors. Postrelapse radiation therapy among previously nonirradiated SJYC07 patients was associated with long-term survival. Reirradiation was only temporizing for SJMB03 patients. Among 127 patients with patient-matched tumor pairs, 9 (7%) experienced subsequent nonmedulloblastoma CNS malignancies. Subgroup (96%) and subtype (80%) stabilities were largely maintained among the remainder. Rare subgroup divergence was observed from Group 4 to Group 3 tumors, which is coincident with genetic alterations involving MYC, MYCN, and FBXW7. Subgroup-specific patterns of alteration were identified for driver genes and chromosome arms. CONCLUSION: Clinical behavior of relapsed medulloblastoma must be contextualized in terms of up-front therapies and molecular classifications. Group 4 tumors exhibit slower biological progression. Utility of radiation at relapse is dependent on patient age and prior treatments. Degree and patterns of molecular conservation at relapse vary by subgroup. Relapse tissue enables verification of molecular targets and identification of occult secondary malignancies.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Neoplasms/genetics , DNA Methylation , Medulloblastoma/genetics , Neoplasm Recurrence, Local , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/therapy , Child , Child, Preschool , Clinical Trials as Topic , Disease Progression , Epigenome , Epigenomics , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Medulloblastoma/mortality , Medulloblastoma/secondary , Medulloblastoma/therapy , Retreatment , Time Factors , Treatment Outcome
6.
Sci Rep ; 8(1): 1032, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348602

ABSTRACT

Astroblastomas are rare brain tumours which predominate in children and young adults, and have a controversial claim as a distinct entity, with no established WHO grade. Reports suggest a better outcome than high grade gliomas, though they frequently recur. Recently, they have been described to overlap with a newly-discovered group of tumours described as'high grade neuroepithelial tumour with MN1 alteration' (CNS HGNET-MN1), defined by global methylation patterns and strongly associated with gene fusions targeting MN1. We have studied a unique case of astroblastoma arising in a 6 year-old girl, with multiple recurrences over a period of 10 years, with the pathognomonic MN1:BEND2 fusion. Exome sequencing allowed for a phylogenetic reconstruction of tumour evolution, which when integrated with clinical, pathological and radiological data provide for a detailed understanding of disease progression, with initial treatment driving tumour dissemination along four distinct trajectories. Infiltration of distant sites was associated with a later genome doubling, whilst there was evidence of convergent evolution of different lesions acquiring distinct alterations targeting NF-κB. These data represent an unusual opportunity to understand the evolutionary history of a highly recurrent childhood brain tumour, and provide novel therapeutic targets for astroblastoma/CNS HGNET-MN1.


Subject(s)
Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/pathology , Oncogene Proteins, Fusion/genetics , Tumor Suppressor Proteins/genetics , Age Factors , Child , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Neoplasm Grading , Neoplasms, Neuroepithelial/diagnostic imaging , Oncogene Proteins, Fusion/metabolism , Recurrence , Trans-Activators , Translocation, Genetic , Tumor Suppressor Proteins/metabolism , Exome Sequencing
7.
Int J Radiat Oncol Biol Phys ; 93(1): 47-53, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26279023

ABSTRACT

PURPOSE: The treatment of children with embryonal brain tumors (EBT) includes craniospinal irradiation (CSI). There are limited data regarding the effect of CSI on pulmonary function. METHODS: Protocol SJMB03 enrolled patients 3 to 21 years of age with EBT. Pulmonary function tests (PFTs) (forced expiratory volume in 1 second [FEV1] and forced vital capacity [FVC] by spirometry, total lung capacity [TLC] by nitrogen washout or plethysmography, and diffusing capacity of the lung for carbon monoxide corrected for hemoglobin [DLCO(corr)]) were obtained. Differences between PFTs obtained immediately after the completion of CSI and 24 or 60 months after the completion of treatment (ACT) were compared using exact Wilcoxon signed-rank tests and repeated-measures models. RESULTS: Between June 24, 2003, and March 1, 2010, 303 eligible patients (spine dose: ≤ 2345 cGy, 201; >2345 cGy, 102; proton beam, 20) were enrolled, 260 of whom had at least 1 PFT. The median age at diagnosis was 8.9 years (range, 3.1-20.4 years). The median thoracic spinal radiation dose was 23.4 Gy (interquartile range [IQR], 23.4-36.0 Gy). The median cyclophosphamide dose was 16.0 g/m(2) (IQR, 15.7-16.0 g/m(2)). At 24 and 60 months ACT, DLCO(corr) was <75% predicted in 23% (27/118) and 25% (21/84) of patients, FEV1 was <80% predicted in 20% (34/170) and 29% (32/109) of patients, FVC was <80% predicted in 27% (46/172) and 28% (30/108) of patients, and TLC was <75% predicted in 9% (13/138) and 11% (10/92) of patients. DLCO(corr) was significantly decreased 24 months ACT (median difference [MD] in % predicted, 3.00%; P = .028) and 60 months ACT (MD in % predicted, 6.00%; P = .033) compared with the end of radiation therapy. These significant decreases in DLCO(corr) were also observed in repeated-measures models (P = .011 and P = .032 at 24 and 60 months ACT, respectively). CONCLUSIONS: A significant minority of EBT survivors experience PFT deficits after CSI. Continued monitoring of this cohort is planned.


Subject(s)
Brain Neoplasms/radiotherapy , Craniospinal Irradiation , Lung/radiation effects , Neoplasms, Germ Cell and Embryonal/radiotherapy , Adolescent , Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Child , Child, Preschool , Cyclophosphamide/administration & dosage , Humans , Lung/physiopathology , Neoplasms, Germ Cell and Embryonal/drug therapy , Respiratory Function Tests , Young Adult
8.
Exp Hematol Oncol ; 2(1): 26, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24219920

ABSTRACT

BACKGROUND: Resistance to radiation treatment remains a major clinical problem for patients with brain cancer. Medulloblastoma is the most common malignant brain tumor of childhood, and occurs in the cerebellum. Though radiation treatment has been critical in increasing survival rates in recent decades, the presence of resistant cells in a substantial number of medulloblastoma patients leads to relapse and death. METHODS: Using the established medulloblastoma cell lines UW228 and Daoy, we developed a novel model system to enrich for and study radiation tolerant cells early after radiation exposure. Using fluorescence-activated cell sorting, dead cells and cells that had initiated apoptosis were removed, allowing surviving cells to be investigated before extensive proliferation took place. RESULTS: Isolated surviving cells were tumorigenic in vivo and displayed elevated levels of ABCG2, an ABC transporter linked to stem cell behavior and drug resistance. Further investigation showed another family member, ABCA1, was also elevated in surviving cells in these lines, as well as in early passage cultures from pediatric medulloblastoma patients. We discovered that the multi-ABC transporter inhibitors verapamil and reserpine sensitized cells from particular patients to radiation, suggesting that ABC transporters have a functional role in cellular radiation protection. Additionally, verapamil had an intrinsic anti-proliferative effect, with transient exposure in vitro slowing subsequent in vivo tumor formation. When expression of key ABC transporter genes was assessed in medulloblastoma tissue from 34 patients, levels were frequently elevated compared with normal cerebellum. Analysis of microarray data from independent cohorts (n = 428 patients) showed expression of a number of ABC transporters to be strongly correlated with certain medulloblastoma subtypes, which in turn are associated with clinical outcome. CONCLUSIONS: ABC transporter inhibitors are already being trialed clinically, with the aim of decreasing chemotherapy resistance. Our findings suggest that the inhibition of ABC transporters could also increase the efficacy of radiation treatment for medulloblastoma patients. Additionally, the finding that certain family members are associated with particular molecular subtypes (most notably high ABCA8 and ABCB4 expression in Sonic Hedgehog pathway driven tumors), along with cell membrane location, suggests ABC transporters are worthy of consideration for the diagnostic classification of medulloblastoma.

9.
Pediatr Blood Cancer ; 58(3): 390-4, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21425449

ABSTRACT

BACKGROUND: International studies examining the association between the incidence of childhood cancer and characteristics of the area in which the patient lives have generally reported inconsistent patterns. Area-based differentials in childhood cancer throughout Australia have not been previously published at a national level. PROCEDURE: Population-based information from the Australian Paediatric Cancer Registry was used to identify all children aged 0- to 14-years old diagnosed with invasive cancer or intracranial and intraspinal tumors of benign or uncertain behavior between 1996 and 2006. Age-standardized incidence rates per million children per year and the corresponding incidence rate ratios were calculated, categorized by remoteness of residence and an area-based index of socioeconomic disadvantage. Results were also stratified by the most common types of childhood cancer. RESULTS: There was a significant, decreasing gradient in the incidence of childhood cancer as remoteness of residence increased. Children living in remote or very remote areas were 21% less likely to be diagnosed with cancer compared to children in major cities, mainly due to differences in the incidence of leukemias and lymphomas. This differential was no longer significant when only non-Indigenous children were considered. No clear relationship was found between incidence and socioeconomic status (SES) in contrast to similar earlier studies. CONCLUSIONS: The findings by remoteness of residence are consistent with the lower incidence rates of cancer that are typically associated with Indigenous Australians. There is also a suggestion that the etiological factors associated with childhood leukemia and SES may have altered over time.


Subject(s)
Health Status Disparities , Native Hawaiian or Other Pacific Islander , Neoplasms/ethnology , Poverty Areas , Rural Health , Adolescent , Australia/epidemiology , Child , Child, Preschool , Female , Humans , Incidence , Infant , Infant, Newborn , Male , Native Hawaiian or Other Pacific Islander/statistics & numerical data , Regression Analysis , Socioeconomic Factors
10.
Pediatr Blood Cancer ; 56(7): 1055-61, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21298769

ABSTRACT

BACKGROUND: ANZCCSG BabyBrain99 is a trial of intensive systemic chemotherapy with dual stem cell supported treatment, second look surgery and involved field radiation for children less than four years of age with malignant central nervous system tumours. PROCEDURE: Following primary resection, treatment included two courses of cisplatin and oral etoposide, a third course of mobilising chemotherapy (vincristine, etoposide, cyclophosphamide) with stem cell harvest, followed by intensive stem cell supported chemotherapy with high dose cyclophosphamide, etoposide and vincristine. Children were evaluated for second resection before proceeding to a second stem cell supported consolidation therapy consisting of melphalan and carboplatin. Patients then received involved field radiation therapy. RESULTS: Thirty three children with a range of diagnoses were enrolled. Nine percent of children had metastatic disease at diagnosis. Eighteen children completed treatment including irradiation. At the end of induction the event free survival was 70% (54-86). Forty eight percent of children had a complete response, 18% had stable disease and 3% had a partial response. Five year overall survival was 40% (22-56) and event free survival was 33% (17-50). Children in whom a complete resection were achieved had a significantly superior outcome compared to those children without a complete resection, 5 year EFS 60% (45-75), as compared to 22% (13-30), P-value <0.05. CONCLUSIONS: BabyBrain99 confirms that intensive stem cell supported chemotherapy can be safely administered to infants with CNS tumours however overall prognosis remains poor. Importantly, the study reinforces a complete surgical resection as an important favourable prognostic indicator. Pediatr Blood Cancer 2011;56:1055-1061. © 2011 Wiley-Liss, Inc.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Glioma/therapy , Spinal Neoplasms/therapy , Brain Neoplasms/pathology , Carboplatin/administration & dosage , Child, Preschool , Cisplatin/administration & dosage , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Etoposide/administration & dosage , Feasibility Studies , Female , Follow-Up Studies , Glioma/secondary , Humans , Infant , Male , Melphalan/administration & dosage , Radiotherapy Dosage , Remission Induction , Second-Look Surgery , Spinal Neoplasms/secondary , Stem Cell Transplantation , Survival Rate , Transplantation, Autologous , Treatment Outcome , Vincristine/administration & dosage
11.
Neuro Oncol ; 6(3): 236-46, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15279716

ABSTRACT

We conducted a phase 1 study of 9 pediatric patients with recurrent brain tumors using monocyte-derived dendritic cells pulsed with tumor RNA to produce antitumor vaccine (DCRNA) preparations. The objectives of this study included (1) establishing safety and feasibility and (2) measuring changes in general, antigen-specific, and tumor-specific immune responses after DCRNA. Dendritic cells were derived from freshly isolated monocytes after 7 days of culture with IL-4 and granulocyte-macrophage colony-stimulating factor, pulsed with autologous tumor RNA, and then cryopreserved. Patients received at least 3 vaccines, each consisting of an intravenous and an intradermal administration at biweekly intervals. The study showed that this method for producing and administering DCRNA from a single leukapheresis product was both feasible and safe in this pediatric brain tumor population. Immune function at the time of enrollment into the study was impaired in all patients tested. While humoral responses to recall antigens (diphtheria and tetanus) were intact in all patients, cellular responses to mitogen and recall antigens were below normal. Following DCRNA vaccine, 2 of 7 patients showed stable clinical disease and 1 of 7 showed a partial response. Two of 7 patients who were tested showed a tumor-specific immune response to DCRNA. This study showed that DCRNA vaccines are both safe and feasible in children with tumors of the central nervous system with a single leukapheresis.


Subject(s)
Brain Neoplasms/drug therapy , Cancer Vaccines/administration & dosage , Dendritic Cells/metabolism , Immunotherapy, Adoptive/methods , Monocytes/metabolism , RNA, Neoplasm/administration & dosage , Adolescent , Adult , Antibodies, Neoplasm/biosynthesis , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Child , Dendritic Cells/immunology , Female , Humans , Immunity, Cellular , Immunotherapy, Adoptive/adverse effects , Interleukin-10/biosynthesis , Lymphocyte Count , Male , Monocytes/immunology , RNA, Neoplasm/adverse effects , RNA, Neoplasm/immunology , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...