Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Pediatr Hematol Oncol ; 46(5): 262-271, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38857189

ABSTRACT

Precision oncology incorporates comprehensive genomic profiling into the individualized clinical care of pediatric cancer patients. In recent years, comprehensive pan-cancer analyses have led to the successful implementation of genomics-based pediatric trials and accelerated approval of novel targeted agents. In addition, disease-specific studies have resulted in molecular subclassification of myriad cancer types with subsequent tailoring of treatment intensity based on the patient's prognostic factors. This review discusses the progress of the field and highlights developments that are leading to more personalized cancer care and improved patient outcomes. Increased understanding of the evolution of precision oncology over recent decades emphasizes the tremendous impact of improved genomic applications. New technologies and improved diagnostic modalities offer further promise for future advancements within the field.


Subject(s)
Medical Oncology , Neoplasms , Precision Medicine , Humans , Precision Medicine/methods , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/diagnosis , Child , Medical Oncology/methods , Medical Oncology/trends , Genomics/methods
2.
J Clin Oncol ; : JCO2302019, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771986

ABSTRACT

PURPOSE: T cells modified with chimeric antigen receptors (CARTs) have demonstrated efficacy for hematologic malignancies; however, benefit for patients with CNS tumors has been limited. To enhance T cell activity against GD2+ CNS malignancies, we modified GD2-directed CART cells (GD2.CARTs) with a constitutively active interleukin (IL)-7 receptor (C7R-GD2.CARTs). METHODS: Patients age 1-21 years with H3K27-altered diffuse midline glioma (DMG) or other recurrent GD2-expressing CNS tumors were eligible for this phase I trial (ClinicalTrials.gov identifier: NCT04099797). All subjects received standard-of-care adjuvant radiation therapy or chemotherapy before study enrollment. The first treatment cohort received GD2.CARTs alone (1 × 107 cells/m2), and subsequent cohorts received C7R-GD2.CARTs at two dose levels (1 × 107 cells/m2; 3 × 107 cells/m2). Standard lymphodepletion with cyclophosphamide and fludarabine was included at all dose levels. RESULTS: Eleven patients (age 4-18 years) received therapy without dose-limiting toxicity. The GD2.CART cohort did not experience toxicity, but had disease progression after brief improvement of residual neurologic deficits (≤3 weeks). The C7R-GD2.CART cohort developed grade 1 tumor inflammation-associated neurotoxicity in seven of eight (88%) cases, controllable with anakinra. Cytokine release syndrome was observed in six of eight (75%, grade 1 in all but one patient) and associated with increased circulating IL-6 and IP-10 (P < .05). Patients receiving C7R-GD2.CARTs experienced temporary improvement from baseline neurologic deficits (range, 2 to >12 months), and seven of eight (88%) remained eligible for additional treatment cycles (range 2-4 cycles). Partial responses by iRANO criteria were observed in two of seven (29%) patients with DMG treated by C7R-GD2.CARTs. CONCLUSION: Intravenous GD2.CARTs with and without C7R were well tolerated. Patients treated with C7R-GD2.CARTs exhibited transient improvement of neurologic deficits and increased circulating cytokines/chemokines. Treatment with C7R-GD2.CARTs represents a novel approach warranting further investigation for children with these incurable CNS cancers.

3.
Pediatr Hematol Oncol ; 40(8): 719-738, 2023.
Article in English | MEDLINE | ID: mdl-37366551

ABSTRACT

The potential of circulating tumor DNA (ctDNA) analysis to serve as a real-time "liquid biopsy" for children with central nervous system (CNS) and non-CNS solid tumors remains to be fully elucidated. We conducted a study to investigate the feasibility and potential clinical utility of ctDNA sequencing in pediatric patients enrolled on an institutional clinical genomics trial. A total of 240 patients had tumor DNA profiling performed during the study period. Plasma samples were collected at study enrollment from 217 patients and then longitudinally from a subset of patients. Successful cell-free DNA extraction and quantification occurred in 216 of 217 (99.5%) of these initial samples. Twenty-four patients were identified whose tumors harbored 30 unique variants that were potentially detectable on a commercially-available ctDNA panel. Twenty of these 30 mutations (67%) were successfully detected by next-generation sequencing in the ctDNA from at least one plasma sample. The rate of ctDNA mutation detection was higher in patients with non-CNS solid tumors (7/9, 78%) compared to those with CNS tumors (9/15, 60%). A higher ctDNA mutation detection rate was also observed in patients with metastatic disease (9/10, 90%) compared to non-metastatic disease (7/14, 50%), although tumor-specific variants were detected in a few patients in the absence of radiographic evidence of disease. This study illustrates the feasibility of incorporating longitudinal ctDNA analysis into the management of relapsed or refractory patients with childhood CNS or non-CNS solid tumors.


Subject(s)
Brain Neoplasms , Circulating Tumor DNA , Humans , Child , Circulating Tumor DNA/genetics , Feasibility Studies , Biomarkers, Tumor , High-Throughput Nucleotide Sequencing , Brain Neoplasms/genetics , Mutation
4.
Surg Neurol Int ; 14: 55, 2023.
Article in English | MEDLINE | ID: mdl-36895253

ABSTRACT

Background: Gangliogliomas (GGs) are rare tumors of the central nervous system composed of neoplastic neural and glial cells and are typically low-grade. Intramedullary spinal anaplastic GGs (AGG) are rare, poorly understood, and often aggressive tumors that can result in widespread progression along the craniospinal axis. Due to the rarity of these tumors, data are lacking to guide clinical and pathologic diagnosis and standard of care treatment. Here, we present a case of pediatric spinal AGG to provide information on our institutional approach to work-up and to highlight unique molecular pathology. Case Description: A 13-year-old female presented with signs of spinal cord compression including right sided hyperreflexia, weakness, and enuresis. Magnetic resonance imaging (MRI) revealed a C3-C5 cystic and solid mass which was treated surgically with osteoplastic laminoplasty and tumor resection. Histopathologic diagnosis was consistent with AGG, and molecular testing identified mutations in H3F3A (K27M), TP53, and NF1. She received adjuvant radiation therapy and her neurological symptoms improved. However, at 6-month follow-up, she developed new symptoms. MRI revealed metastatic recurrence of tumor with leptomeningeal and intracranial spread. Conclusion: Primary spinal AGGs are rare tumors, but a growing body of literature shows some trends that may improve diagnosis and management. These tumors generally present in adolescence and early adulthood with motor/sensory impairment and other spinal cord symptoms. They are most commonly treated by surgical resection but frequently recur due to their aggressive nature. Further reports of these primary spinal AGGs along with characterization of their molecular profile will be important in developing more effective treatments.

5.
Pediatr Blood Cancer ; 69(11): e29859, 2022 11.
Article in English | MEDLINE | ID: mdl-35713195

ABSTRACT

BACKGROUND: The association of childhood cancer with Lynch syndrome is not established compared with the significant pediatric cancer risk in recessive constitutional mismatch repair deficiency syndrome (CMMRD). PROCEDURE: We describe the clinical features, germline analysis, and tumor genomic profiling of patients with Lynch syndrome among patients enrolled in pediatric cancer genomic studies. RESULTS: There were six of 773 (0.8%) pediatric patients with solid tumors identified with Lynch syndrome, defined as a germline heterozygous pathogenic variant in one of the mismatch repair (MMR) genes (three with MSH6, two with MLH1, and one with MSH2). Tumor analysis demonstrated evidence for somatic second hits and/or increased tumor mutation burden in three of four patients with available tumor with potential implications for therapy and identification of at-risk family members. Only one patient met current guidelines for pediatric cancer genetics evaluation at the time of tumor diagnosis. CONCLUSION: Approximately 1% of children with cancer have Lynch syndrome, which is missed with current referral guidelines, suggesting the importance of adding MMR genes to tumor and hereditary pediatric cancer panels. Tumor analysis may provide the first suggestion of an underlying cancer predisposition syndrome and is useful in distinguishing between Lynch syndrome and CMMRD.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Brain Neoplasms , Child , Colorectal Neoplasms , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Germ-Line Mutation , Humans , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Neoplastic Syndromes, Hereditary
8.
Pediatr Blood Cancer ; 68(10): e29178, 2021 10.
Article in English | MEDLINE | ID: mdl-34133064

ABSTRACT

BACKGROUND: Patients with chiasmatic-hypothalamic low-grade glioma (CHLGG) have frequent MRIs with gadolinium-based contrast agents (GBCA) for disease monitoring. Cumulative gadolinium deposition in the brains of children is a potential concern. The purpose of this study is to evaluate whether MRI with GBCA is necessary for determining radiographic tumor progression in children with CHLGG. METHODS: Children who were treated for progressive CHLGG from 2005 to 2019 at Texas Children's Cancer Center were identified. Pre- and post-contrast MRI sequences were separately reviewed by one neuroradiologist who was blinded to the clinical course. Three dimensional measurements and tumor characteristics were evaluated. Radiographic progression was defined as a 25% increase in size (product of two largest dimensions) compared with baseline or best response after initiation of therapy. RESULTS: A total of 28 patients with progressive CHLGG were identified with a total of 683 MRIs with GBCA reviewed (mean 24 MRIs/patient; range, 11-43 MRIs). Radiographic progression was observed 92 times, 91 (99%) on noncontrast and 90 (98%) on contrast imaging. Sixty-seven progressions necessitating management changes were identified in all (100%) noncontrast sequences and 66 (99%) contrast sequences. Tumor growth > 2 mm in any dimension was identified in 184/187 (98%) noncontrast and 181/187 (97%) with contrast imaging. Metastatic tumors were better visualized on contrast imaging in 4/7 (57%). CONCLUSION: MRI without GBCA effectively identifies patients with progressive disease. When imaging children with CHLGG, eliminating GBCA should be considered unless monitoring patients with metastatic disease.


Subject(s)
Gadolinium , Glioma , Brain/diagnostic imaging , Child , Contrast Media , Glioma/diagnostic imaging , Humans , Magnetic Resonance Imaging , Retrospective Studies
9.
Clin Perinatol ; 48(1): 53-70, 2021 03.
Article in English | MEDLINE | ID: mdl-33583507

ABSTRACT

Retinoblastoma is the most common ocular malignancy of childhood. With an estimated 300 cases annually in the United States, retinoblastoma is nevertheless considered a rare tumor. Although retinoblastoma primarily affects younger children, diagnosis during the neonatal age range is less common. However, an understanding of patients at risk is critical for appropriate screening. Early detection and treatment by a multidisciplinary specialty team maximizes the chance for survival and ocular/vision salvage while minimizing treatment-related toxicity. Testing for alterations in the RB1 gene has become standard practice, and informs screening and genetic counseling recommendations for patients and their families.


Subject(s)
Retinal Neoplasms , Retinoblastoma , Child , Early Diagnosis , Genetic Counseling , Humans , Infant, Newborn , Mass Screening , Retinal Neoplasms/diagnosis , Retinal Neoplasms/epidemiology , Retinal Neoplasms/genetics , Retinoblastoma/diagnosis , Retinoblastoma/epidemiology , Retinoblastoma/genetics
10.
Article in English | MEDLINE | ID: mdl-31624068

ABSTRACT

Ultra-hypermutation (>100 mutations/Mb) is rare in childhood cancer genomes and has been primarily reported in patients with constitutional mismatch repair deficiency (CMMRD) caused by biallelic germline mismatch repair (MMR) gene mutations. We report a 5-yr-old child with classic clinical features of CMMRD and an ultra-hypermutated medulloblastoma with retained MMR protein expression and absence of germline MMR mutations. Mutational signature analysis of tumor panel sequencing data revealed a canonical DNA polymerase-deficiency-associated signature, prompting further genetic testing that uncovered a germline POLE p.A456P missense variant, which has previously been reported as a recurrent somatic driver mutation in cancers. This represents the earliest known onset of malignancy in a patient with a germline mutation in the POLE proofreading polymerase. The clinical features in this child, virtually indistinguishable from those of CMMRD, suggest that polymerase-proofreading deficiency should be considered in the differential diagnosis of CMMRD patients with retained MMR function.


Subject(s)
Brain Neoplasms/genetics , Colorectal Neoplasms/genetics , DNA Polymerase II/genetics , Medulloblastoma/genetics , Neoplastic Syndromes, Hereditary/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Brain Neoplasms/metabolism , Cerebellar Neoplasms , Child, Preschool , Colorectal Neoplasms/metabolism , DNA Mismatch Repair/genetics , DNA Mutational Analysis , DNA Polymerase II/metabolism , DNA-Binding Proteins/genetics , Female , Genetic Predisposition to Disease , Genetic Testing , Germ Cells/metabolism , Germ-Line Mutation/genetics , Humans , Medulloblastoma/metabolism , Mutation , Neoplastic Syndromes, Hereditary/metabolism , Phenotype , Poly-ADP-Ribose Binding Proteins/metabolism
11.
Clin Cancer Res ; 24(9): 2159-2170, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29463553

ABSTRACT

Purpose: Pediatric glioblastoma multiforme (pGBM) is a highly aggressive tumor in need of novel therapies. Our objective was to demonstrate the therapeutic efficacy of MLN8237 (alisertib), an orally available selective inhibitor of Aurora A kinase (AURKA), and to evaluate which in vitro model system (monolayer or neurosphere) can predict therapeutic efficacy in vivoExperimental Design: AURKA mRNA expressions were screened with qRT-PCR. In vitro antitumor effects were examined in three matching pairs of monolayer and neurosphere lines established from patient-derived orthotopic xenograft (PDOX) models of the untreated (IC-4687GBM), recurrent (IC-3752GBM), and terminal (IC-R0315GBM) tumors, and in vivo therapeutic efficacy through log rank analysis of survival times in two models (IC-4687GBM and IC-R0315GBM) following MLN8237 treatment (30 mg/kg/day, orally, 12 days). Drug concentrations in vivo and mechanism of action and resistance were also investigated.Results: AURKA mRNA overexpression was detected in 14 pGBM tumors, 10 PDOX models, and 6 cultured pGBM lines as compared with 11 low-grade gliomas and normal brains. MLN8237 penetrated into pGBM xenografts in mouse brains. Significant extension of survival times were achieved in IC-4687GBM of which both neurosphere and monolayer were inhibited in vitro, but not in IC-R0315GBM of which only neurosphere cells responded (similar to IC-3752GBM). Apoptosis-mediated MLN8237 induced cell death, and the presence of AURKA-negative and CD133+ cells appears to have contributed to in vivo therapy resistance.Conclusions: MLN8237 successfully targeted AURKA in a subset of pGBMs. Our data suggest that combination therapy should aim at AURKA-negative and/or CD133+ pGBM cells to prevent tumor recurrence. Clin Cancer Res; 24(9); 2159-70. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Biomarkers , Biomarkers, Tumor , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Child , Child, Preschool , Disease Models, Animal , Female , Flow Cytometry , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Immunohistochemistry , Immunophenotyping , Male , Neoplasm Grading , Xenograft Model Antitumor Assays
12.
Pediatr Hematol Oncol ; 34(4): 254-259, 2017 May.
Article in English | MEDLINE | ID: mdl-29040023

ABSTRACT

While clinical and radiographic responses to agents targeting the mitogen-activated protein kinases (MAPK) pathway have been repor-ted in pediatric low-grade gliomas (LGG), early phase trials indicate refractoriness to these medications in some of these patients. We report a patient with disseminated LGG with the BRAFV600E mutation, which was refractory to selumetinib, a MEK inhibitor, but subsequently showed immediate clinical and radiographic response to dabrafenib, a BRAF inhibitor, with sustained effect for 9 months prior to clinical progression. In LGGs, treatment resistance to one agent targeting the MAPK pathway might not imply refractoriness to other agents targeting this pathway.


Subject(s)
Benzimidazoles/administration & dosage , Chemoradiotherapy , Glioma , Imidazoles/administration & dosage , Mutation, Missense , Optic Nerve Neoplasms , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf , Amino Acid Substitution , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Glioma/therapy , Humans , Infant , Male , Optic Nerve Neoplasms/enzymology , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/pathology , Optic Nerve Neoplasms/therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
13.
Pediatr Dermatol ; 34(3): 337-341, 2017 May.
Article in English | MEDLINE | ID: mdl-28523881

ABSTRACT

Vemurafenib is increasingly being used to treat nonmelanoma tumors that are positive for the BRAF V600E mutation. We report three children who presented with panniculitis induced by vemurafenib while undergoing treatment for central nervous system tumors and review the literature.


Subject(s)
Antineoplastic Agents/adverse effects , Central Nervous System Neoplasms/drug therapy , Indoles/adverse effects , Panniculitis/chemically induced , Sulfonamides/adverse effects , Adolescent , Child , Humans , Male , Vemurafenib
14.
Cold Spring Harb Mol Case Stud ; 2(5): a001057, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27626068

ABSTRACT

The integration of genome-scale studies such as whole-exome sequencing (WES) into the clinical care of children with cancer has the potential to provide insight into the genetic basis of an individual's cancer with implications for clinical management. This report describes the results of clinical tumor and germline WES for a patient with a rare tumor diagnosis, rosette-forming glioneuronal tumor of the fourth ventricle (RGNT). Three pathogenic gene alterations with implications for clinical care were identified: somatic activating hotspot mutations in FGFR1 (p.N546K) and PIK3CA (p.H1047R) and a germline pathogenic variant in PTPN11 (p.N308S) diagnostic for Noonan syndrome. The molecular landscape of RGNT is not well-described, but these data are consistent with prior observations regarding the importance of the interconnected MAPK and PI3K/AKT/mTOR signaling pathways in this rare tumor. The co-occurrence of FGFR1, PIK3CA, and PTPN11 alterations provides further evidence for consideration of RGNT as a distinct molecular entity from pediatric low-grade gliomas and suggests potential therapeutic strategies for this patient in the event of tumor recurrence as novel agents targeting these pathways enter pediatric clinical trials. Although RGNT has not been definitively linked with cancer predisposition syndromes, two prior cases have been reported in patients with RASopathies (Noonan syndrome and neurofibromatosis type 1 [NF1]), providing an additional link between these tumors and the mitogen-activated protein kinase (MAPK) signaling pathway. In summary, this case provides an example of the potential for genome-scale sequencing technologies to provide insight into the biology of rare tumors and yield both tumor and germline results of potential relevance to patient care.

15.
Oncology (Williston Park) ; 30(5): 411-23, 2016 May.
Article in English | MEDLINE | ID: mdl-27188671

ABSTRACT

Recent advances in genome-scale sequencing methods have resulted in a significant increase in our understanding of the biology of human cancers. When applied to pediatric central nervous system (CNS) tumors, these remarkable technological breakthroughs have facilitated the molecular characterization of multiple tumor types, provided new insights into the genetic basis of these cancers, and prompted innovative strategies that are changing the management paradigm in pediatric neuro-oncology. Genomic tests have begun to affect medical decision making in a number of ways, from delineating histopathologically similar tumor types into distinct molecular subgroups that correlate with clinical characteristics, to guiding the addition of novel therapeutic agents for patients with high-risk or poor-prognosis tumors, or alternatively, reducing treatment intensity for those with a favorable prognosis. Genomic sequencing has also had a significant impact on translational research strategies in pediatric CNS tumors, resulting in wide-ranging applications that have the potential to direct the rational preclinical screening of novel therapeutic agents, shed light on tumor heterogeneity and evolution, and highlight differences (or similarities) between pediatric and adult CNS tumors. Finally, in addition to allowing the identification of somatic (tumor-specific) mutations, the analysis of patient-matched constitutional (germline) DNA has facilitated the detection of pathogenic germline alterations in cancer genes in patients with CNS tumors, with critical implications for genetic counseling and tumor surveillance strategies for children with familial predisposition syndromes. As our understanding of the molecular landscape of pediatric CNS tumors continues to advance, innovative applications of genomic sequencing hold significant promise for further improving the care of children with these cancers.


Subject(s)
Biomarkers, Tumor/genetics , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/genetics , DNA Mutational Analysis , Genomics/methods , Mutation , Age Factors , Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/mortality , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Genetic Predisposition to Disease , Humans , Infant , Molecular Targeted Therapy , Patient Selection , Phenotype , Precision Medicine , Predictive Value of Tests , Prognosis
16.
JAMA Oncol ; 2(5): 616-624, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26822237

ABSTRACT

IMPORTANCE: Whole-exome sequencing (WES) has the potential to reveal tumor and germline mutations of clinical relevance, but the diagnostic yield for pediatric patients with solid tumors is unknown. OBJECTIVE: To characterize the diagnostic yield of combined tumor and germline WES for children with solid tumors. DESIGN: Unselected children with newly diagnosed and previously untreated central nervous system (CNS) and non-CNS solid tumors were prospectively enrolled in the BASIC3 study at a large academic children's hospital during a 23-month period from August 2012 through June 2014. Blood and tumor samples underwent WES in a certified clinical laboratory with genetic results categorized on the basis of perceived clinical relevance and entered in the electronic health record. MAIN OUTCOMES AND MEASURES: Clinical categorization of somatic mutations; frequencies of deleterious germline mutations related to patient phenotype and incidental medically-actionable mutations. RESULTS: Of the first 150 participants (80 boys and 70 girls, mean age, 7.4 years), tumor samples adequate for WES were available from 121 patients (81%). Somatic mutations of established clinical utility (category I) were reported in 4 (3%) of 121 patients, with mutations of potential utility (category II) detected in an additional 29 (24%) of 121 patients. CTNNB1 was the gene most frequently mutated, with recurrent mutations in KIT, TSC2, and MAPK pathway genes (BRAF, KRAS, and NRAS) also identified. Mutations in consensus cancer genes (category III) were found in an additional 24 (20%) of 121 tumors. Fewer than half of somatic mutations identified were in genes known to be recurrently mutated in the tumor type tested. Diagnostic germline findings related to patient phenotype were discovered in 15 (10%) of 150 cases: 13 pathogenic or likely pathogenic dominant mutations in adult and pediatric cancer susceptibility genes (including 2 each in TP53, VHL, and BRCA1), 1 recessive liver disorder with hepatocellular carcinoma (TJP2), and 1 renal diagnosis (CLCN5). Incidental findings were reported in 8 (5%) of 150 patients. Most patients harbored germline uncertain variants in cancer genes (98%), pharmacogenetic variants (89%), and recessive carrier mutations (85%). CONCLUSIONS AND RELEVANCE: Tumor and germline WES revealed mutations in a broad spectrum of genes previously implicated in both adult and pediatric cancers. Combined reporting of tumor and germline WES identified diagnostic and/or potentially actionable findings in nearly 40% of newly diagnosed pediatric patients with solid tumors.

17.
Curr Oncol Rep ; 17(10): 47, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26369328

ABSTRACT

Ependymomas are a heterogeneous group of neuroepithelial tumors of children and adults. In pediatric cases, the standard of care has long consisted of neurosurgical resection to the greatest extent acceptable followed by adjuvant involved field irradiation. Complete macroscopic surgical resection has remained the only consistent clinical variable known to improve survival. Adjuvant chemotherapy has yet to predictably affect outcome, possibly due to the molecular heterogeneity of histologically similar tumors. The administration of chemotherapy subsequently remains limited to clinical trials. However, recent comprehensive genomic, transcriptomic, and epigenetic interrogations of ependymomas have uncovered unique molecular characteristics and subtypes that correlated with clinical features such as age, neuroanatomical location, and prognosis. These findings represent a potential paradigm shift and provide a biologic rationale for targeted therapeutic strategies and risk-adapted administration of conventional treatment modalities. In this review, we focus on intracranial WHO grade II and III ependymoma of children and discuss conventional management strategies, followed by recent biologic findings and novel therapeutics currently under investigation.


Subject(s)
Brain Neoplasms/therapy , Chemotherapy, Adjuvant , Ependymoma/therapy , Neurosurgical Procedures/trends , Radiotherapy, Adjuvant , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chemotherapy, Adjuvant/trends , Child , Child, Preschool , Disease-Free Survival , Ependymoma/genetics , Ependymoma/mortality , Ependymoma/pathology , Genetic Markers , Humans , Infant , Prognosis , Radiotherapy, Adjuvant/trends
18.
Mod Pathol ; 28(4): 575-86, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25360585

ABSTRACT

The BCOR-CCNB3 fusion gene, resulting from a chromosome X paracentric inversion, was recently described in translocation-negative 'Ewing-like' sarcomas arising in bone and soft tissue. Genetic subclassification of undifferentiated unclassified sarcomas may potentially offer markers for reproducible diagnosis and substrates for therapy. Using whole transcriptome paired-end RNA sequencing (RNA-seq) we unexpectedly identified BCOR-CCNB3 fusion transcripts in an undifferentiated spindle-cell sarcoma. RNA-seq results were confirmed through direct RT-PCR of tumor RNA and cloning of the genomic breakpoints from tumor DNA. Five additional undifferentiated sarcomas with BCOR-CCNB3 fusions were identified in a series of 42 pediatric and adult unclassified sarcomas. Genomic breakpoint analysis demonstrated unique breakpoint locations in each case at the DNA level even though the resulting fusion mRNA was identical in all cases. All patients with BCOR-CCNB3 sarcoma were males diagnosed in mid childhood (7-13 years of age). Tumors were equally distributed between axial and extra-axial locations. Five of the six tumors were soft-tissue lesions with either predominant spindle-cell morphology or spindle-cell areas interspersed with ovoid to round cells. CCNB3 immunohistochemistry showed strong nuclear positivity in five tumors before oncologic therapy, but was patchy to negative in post-treatment tumor samples. An RT-PCR assay developed to detect the fusion transcript in archival formalin-fixed tissue was positive in all six cases, with high sensitivity and specificity in both pre- and post-treated samples. This study adds to recent reports on the clinicopathologic spectrum of BCOR-CCNB3 fusion-positive sarcomas, a newly emerging entity within the undifferentiated unclassified sarcoma category and describes a simple RT-PCR assay that in conjunction with CCNB3 immunohistochemistry can be useful in diagnosing these tumors.


Subject(s)
Cyclin B/genetics , Oncogene Fusion/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Sarcoma/genetics , Soft Tissue Neoplasms/genetics , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Young Adult
19.
J Med Syst ; 36(4): 2493-503, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21526332

ABSTRACT

Changes in global population and demography, and advances in medicine have led to elderly population growth, creating aging societies from which elderly medical care has evolved. In addition, with the elderly susceptible to chronic diseases, this together with the changing lifestyles of young adults have not only pushed up patient numbers of chronic diseases, but also effected into younger patients. These problems have become the major focus for the health care industry. In response to patient demand and the huge shortage of medical resources, we propose remote healthcare medical information systems that combine patient physiological data acquisition equipment with real-time health care analyses. Since remote health care systems are structured around the Internet, in addition to considering the numerous public systems spread across insecure heterogeneous networks, compatibility among heterogeneous networks will also be another concern. To address the aforementioned issues, mobile agents are adopted. With a mobile agent's characteristics of easy adaptability to heterogeneity and autonomy, the problem of heterogeneous network environments can be tackled. To construct a hierarchical safe access control mechanism for monitoring and control of patient data in order to provide the most appropriate medical treatment, we also propose to use the Chinese Remainder Theorem and discrete logarithm to classify different levels of monitoring staff and hence, to grant permission and access according to their authorized levels. We expect the methods proposed can improve medical care quality and reduce medical resource wastage, while ensuring patient privacy. Finally, security analysis of the system is conducted by simulating a variety of typical attacks, from which it can be concluded that the constructed remote healthcare information system be secure.


Subject(s)
Computer Security , Internet , Medical Records Systems, Computerized , Telecommunications , Algorithms , Confidentiality , Humans
20.
J Med Syst ; 36(3): 1345-57, 2012 Jun.
Article in English | MEDLINE | ID: mdl-20857325

ABSTRACT

Patient records, including doctors' diagnoses of diseases, trace of treatments and patients' conditions, nursing actions, and examination results from allied health profession departments, are the most important medical records of patients in medical systems. With patient records, medical staff can instantly understand the entire medical information of a patient so that, according to the patient's conditions, more accurate diagnoses and more appropriate in-depth treatments can be provided. Nevertheless, in such a modern society with booming information technologies, traditional paper-based patient records have faced a lot of problems, such as lack of uniform formats, low data mobility, slow data transfer, illegible handwritings, enormous and insufficient storage space, difficulty of conservation, being easily damaged, and low transferability. To improve such drawbacks, reduce medical costs, and advance medical quality, paper-based patient records are modified into electronic medical records and reformed into electronic patient records. However, since electronic patient records used in various hospitals are diverse and different, in consideration of cost, it is rather difficult to establish a compatible and complete integrated electronic patient records system to unify patient records from heterogeneous systems in hospitals. Moreover, as the booming of the Internet, it is no longer necessary to build an integrated system. Instead, doctors can instantly look up patients' complete information through the Internet access to electronic patient records as well as avoid the above difficulties. Nonetheless, the major problem of accessing to electronic patient records cross-hospital systems exists in the security of transmitting and accessing to the records in case of unauthorized medical personnels intercepting or stealing the information. This study applies the Mobile Agent scheme to cope with the problem. Since a Mobile Agent is a program, which can move among hosts and automatically disperse arithmetic processes, and moves from one host to another in heterogeneous network systems with the characteristics of autonomy and mobility, decreasing network traffic, reducing transfer lag, encapsulating protocol, availability on heterogeneous platforms, fault-tolerance, high flexibility, and personalization. However, since a Mobile Agent contacts and exchanges information with other hosts or agents on the Internet for rapid exchange and access to medical information, the security is threatened. In order to solve the problem, this study proposes a key management scheme based on Lagrange interpolation formulas and hierarchical management structure to make Mobile Agents a more secure and efficient access control scheme for electronic patient record systems when applied to the access of patients' personal electronic patient records cross hospitals. Meanwhile, with the comparison of security and efficacy analyses being the feasibility of validation scheme and the basis of better efficiency, the security of Mobile Agents in the process of operation can be guaranteed, key management efficacy can be advanced, and the security of the Mobile Agent system can be protected.


Subject(s)
Access to Information , Computer Communication Networks/organization & administration , Computer Security , Electronic Health Records/organization & administration , Algorithms , Internet
SELECTION OF CITATIONS
SEARCH DETAIL
...