Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 84(2): 261-276.e18, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38176414

ABSTRACT

A hallmark of high-risk childhood medulloblastoma is the dysregulation of RNA translation. Currently, it is unknown whether medulloblastoma dysregulates the translation of putatively oncogenic non-canonical open reading frames (ORFs). To address this question, we performed ribosome profiling of 32 medulloblastoma tissues and cell lines and observed widespread non-canonical ORF translation. We then developed a stepwise approach using multiple CRISPR-Cas9 screens to elucidate non-canonical ORFs and putative microproteins implicated in medulloblastoma cell survival. We determined that multiple lncRNA-ORFs and upstream ORFs (uORFs) exhibited selective functionality independent of main coding sequences. A microprotein encoded by one of these ORFs, ASNSD1-uORF or ASDURF, was upregulated, associated with MYC-family oncogenes, and promoted medulloblastoma cell survival through engagement with the prefoldin-like chaperone complex. Our findings underscore the fundamental importance of non-canonical ORF translation in medulloblastoma and provide a rationale to include these ORFs in future studies seeking to define new cancer targets.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Humans , Protein Biosynthesis , Medulloblastoma/genetics , Open Reading Frames/genetics , Cell Survival/genetics , Cerebellar Neoplasms/genetics
2.
Annu Rev Genet ; 53: 483-503, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31794268

ABSTRACT

The human brain contains a vast number of cells and shows extraordinary cellular diversity to facilitate the many cognitive and automatic commands governing our bodily functions. This complexity arises partly from large-scale structural variations in the genome, evolutionary processes to increase brain size, function, and cognition. Not surprisingly given recent technical advances, low-grade gliomas (LGGs), which arise from the glia (the most abundant cell type in the brain), have undergone a recent revolution in their classification and therapy, especially in the pediatric setting. Next-generation sequencing has uncovered previously unappreciated diverse LGG entities, unraveling genetic subgroups and multiple molecular alterations and altered pathways, including many amenable to therapeutic targeting. In this article we review these novel entities, in which oncogenic processes show striking age-related neuroanatomical specificity (highlighting their close interplay with development); the opportunities they provide for targeted therapies, some of which are already practiced at the bedside; and the challenges of implementing molecular pathology in the clinic.


Subject(s)
Brain Neoplasms/genetics , Brain/growth & development , Glioma/genetics , Adult , Age Factors , Brain/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Child , Glioma/diagnosis , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Isocitrate Dehydrogenase/genetics , Molecular Diagnostic Techniques , Mutation , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Receptor, Fibroblast Growth Factor, Type 1/genetics , raf Kinases/genetics
3.
Nature ; 578(7793): 102-111, 2020 02.
Article in English | MEDLINE | ID: mdl-32025015

ABSTRACT

The discovery of drivers of cancer has traditionally focused on protein-coding genes1-4. Here we present analyses of driver point mutations and structural variants in non-coding regions across 2,658 genomes from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium5 of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). For point mutations, we developed a statistically rigorous strategy for combining significance levels from multiple methods of driver discovery that overcomes the limitations of individual methods. For structural variants, we present two methods of driver discovery, and identify regions that are significantly affected by recurrent breakpoints and recurrent somatic juxtapositions. Our analyses confirm previously reported drivers6,7, raise doubts about others and identify novel candidates, including point mutations in the 5' region of TP53, in the 3' untranslated regions of NFKBIZ and TOB1, focal deletions in BRD4 and rearrangements in the loci of AKR1C genes. We show that although point mutations and structural variants that drive cancer are less frequent in non-coding genes and regulatory sequences than in protein-coding genes, additional examples of these drivers will be found as more cancer genomes become available.


Subject(s)
Genome, Human/genetics , Mutation/genetics , Neoplasms/genetics , DNA Breaks , Databases, Genetic , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Humans , INDEL Mutation
4.
Nucleic Acids Res ; 52(5): 2372-2388, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38214234

ABSTRACT

Pediatric high-grade gliomas (pHGG) are devastating and incurable brain tumors with recurrent mutations in histone H3.3. These mutations promote oncogenesis by dysregulating gene expression through alterations of histone modifications. We identify aberrant DNA repair as an independent mechanism, which fosters genome instability in H3.3 mutant pHGG, and opens new therapeutic options. The two most frequent H3.3 mutations in pHGG, K27M and G34R, drive aberrant repair of replication-associated damage by non-homologous end joining (NHEJ). Aberrant NHEJ is mediated by the DNA repair enzyme polynucleotide kinase 3'-phosphatase (PNKP), which shows increased association with mutant H3.3 at damaged replication forks. PNKP sustains the proliferation of cells bearing H3.3 mutations, thus conferring a molecular vulnerability, specific to mutant cells, with potential for therapeutic targeting.


Subject(s)
Brain Neoplasms , Glioma , Histones , Child , Humans , Brain Neoplasms/pathology , DNA Repair/genetics , DNA Repair Enzymes/metabolism , Glioma/pathology , Histones/genetics , Histones/metabolism , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics
5.
Nature ; 576(7785): 112-120, 2019 12.
Article in English | MEDLINE | ID: mdl-31748746

ABSTRACT

The evolutionary processes that drive universal therapeutic resistance in adult patients with diffuse glioma remain unclear1,2. Here we analysed temporally separated DNA-sequencing data and matched clinical annotation from 222 adult patients with glioma. By analysing mutations and copy numbers across the three major subtypes of diffuse glioma, we found that driver genes detected at the initial stage of disease were retained at recurrence, whereas there was little evidence of recurrence-specific gene alterations. Treatment with alkylating agents resulted in a hypermutator phenotype at different rates across the glioma subtypes, and hypermutation was not associated with differences in overall survival. Acquired aneuploidy was frequently detected in recurrent gliomas and was characterized by IDH mutation but without co-deletion of chromosome arms 1p/19q, and further converged with acquired alterations in the cell cycle and poor outcomes. The clonal architecture of each tumour remained similar over time, but the presence of subclonal selection was associated with decreased survival. Finally, there were no differences in the levels of immunoediting between initial and recurrent gliomas. Collectively, our results suggest that the strongest selective pressures occur during early glioma development and that current therapies shape this evolution in a largely stochastic manner.


Subject(s)
Glioma/genetics , Adult , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 19 , Disease Progression , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Polymorphism, Single Nucleotide , Recurrence
6.
Nature ; 560(7718): 325-330, 2018 08.
Article in English | MEDLINE | ID: mdl-30089904

ABSTRACT

Human cancer cell lines are the workhorse of cancer research. Although cell lines are known to evolve in culture, the extent of the resultant genetic and transcriptional heterogeneity and its functional consequences remain understudied. Here we use genomic analyses of 106 human cell lines grown in two laboratories to show extensive clonal diversity. Further comprehensive genomic characterization of 27 strains of the common breast cancer cell line MCF7 uncovered rapid genetic diversification. Similar results were obtained with multiple strains of 13 additional cell lines. Notably, genetic changes were associated with differential activation of gene expression programs and marked differences in cell morphology and proliferation. Barcoding experiments showed that cell line evolution occurs as a result of positive clonal selection that is highly sensitive to culture conditions. Analyses of single-cell-derived clones demonstrated that continuous instability quickly translates into heterogeneity of the cell line. When the 27 MCF7 strains were tested against 321 anti-cancer compounds, we uncovered considerably different drug responses: at least 75% of compounds that strongly inhibited some strains were completely inactive in others. This study documents the extent, origins and consequences of genetic variation within cell lines, and provides a framework for researchers to measure such variation in efforts to support maximally reproducible cancer research.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Evolution, Molecular , Genetic Variation/genetics , Genomic Instability/genetics , Transcription, Genetic/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cell Shape , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Genetic Variation/drug effects , Genomic Instability/drug effects , Humans , MCF-7 Cells , Reproducibility of Results
8.
Curr Opin Neurol ; 35(6): 772-778, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36226704

ABSTRACT

PURPOSE OF REVIEW: The last few decades have seen an explosion in our understanding of the molecular drivers of childhood brain tumours. These insights have opened the possibility for precision medicine approaches for some tumour types. However, a different spectrum of tumours is more likely to occur in infants and young children, who face additional therapeutic challenges. This review focuses on recent advances in molecular genetics of common infant brain tumours and their implication for diagnosis, prognostication and utilization of precision oncology approaches. RECENT FINDINGS: Infant tumours have different biology and outcomes than similar tumours in older children and adults. For low-grade gliomas, targeted MAPK inhibition is well tolerated and likely efficacious. In high-grade gliomas, common tyrosine kinase alterations offer compelling targets for inhibition that are currently being evaluated. Paediatric-specific sequencing and methylation analysis offer insights into the driving biology of infant medulloblastoma, atypical teratoid rhabdoid tumours, embryonal tumours with multilayered rosettes, ependymoma and choroid plexus tumours, with molecular subgrouping shedding insights into distinct driving biology and clinical outcomes. SUMMARY: Infant brain tumours are rare and heterogenous, with overall poor outcomes. Advances in molecular genetics have been incorporated into their diagnostic criteria and allow for accurate subgrouping and improved prognostication. The utilization of targeted agents appears beneficial for many low-grade gliomas and a subset of high-grade gliomas, but further research is urgently needed to improve outcomes for other tumour entities.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Glioma , Infant , Adult , Child , Humans , Child, Preschool , Precision Medicine , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Glioma/pathology , Molecular Biology
9.
Trends Immunol ; 40(8): 748-761, 2019 08.
Article in English | MEDLINE | ID: mdl-31229353

ABSTRACT

Pediatric brain tumors are the leading cause of childhood cancer-related death. Immunotherapy is a powerful new approach for treating some refractory cancers; applying this 'fourth pillar' of cancer treatment to pediatric brain tumors is an exciting but challenging prospect. This review offers new perspectives on moving towards successful immunotherapy for pediatric brain tumors, focusing on pediatric high-grade glioma (HGG), a subgroup with universally poor outcomes. We cover chimeric antigen receptor T cell (CAR-T) therapy, vaccine therapy, and checkpoint inhibition in this context, and focus on the need for intimately understanding the growing brain and its immune system. We highlight the challenges associated with the application of immunotherapy in pediatric neuro-oncology, as well as the tissue-specific challenges to be overcome, to achieve improved outcomes.


Subject(s)
Brain Neoplasms/therapy , Immunotherapy , Age Factors , Animals , Antineoplastic Agents, Immunological , Biomarkers, Tumor , Brain Neoplasms/etiology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Child , Disease Management , Disease Susceptibility , Humans , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Molecular Targeted Therapy , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
10.
Genome Res ; 28(4): 581-591, 2018 04.
Article in English | MEDLINE | ID: mdl-29535149

ABSTRACT

Structural variants (SVs), including small insertion and deletion variants (indels), are challenging to detect through standard alignment-based variant calling methods. Sequence assembly offers a powerful approach to identifying SVs, but is difficult to apply at scale genome-wide for SV detection due to its computational complexity and the difficulty of extracting SVs from assembly contigs. We describe SvABA, an efficient and accurate method for detecting SVs from short-read sequencing data using genome-wide local assembly with low memory and computing requirements. We evaluated SvABA's performance on the NA12878 human genome and in simulated and real cancer genomes. SvABA demonstrates superior sensitivity and specificity across a large spectrum of SVs and substantially improves detection performance for variants in the 20-300 bp range, compared with existing methods. SvABA also identifies complex somatic rearrangements with chains of short (<1000 bp) templated-sequence insertions copied from distant genomic regions. We applied SvABA to 344 cancer genomes from 11 cancer types and found that short templated-sequence insertions occur in ∼4% of all somatic rearrangements. Finally, we demonstrate that SvABA can identify sites of viral integration and cancer driver alterations containing medium-sized (50-300 bp) SVs.


Subject(s)
Genome, Human/genetics , Genomic Structural Variation/genetics , Genomics , INDEL Mutation/genetics , Databases, Genetic , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, DNA , Sequence Deletion/genetics , Software , Virus Integration/genetics
11.
Curr Opin Pediatr ; 33(1): 26-32, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33278109

ABSTRACT

PURPOSE OF REVIEW: Recent genetic and molecular findings have impacted the diagnosis, prognosis, and in some instances, treatment strategies for children with pediatric central nervous system tumors. Herein, we review the most up-to-date molecular findings and how they have impacted tumor classification and clinical care. RECENT FINDINGS: It is now recognized that aberrations of the mitogen-activated protein kinase pathway are present in the majority of pediatric low-grade glioma. Also, there has been the identification of recurrent histone H3 K27M mutations in diffuse intrinsic pontine and other midline gliomas. Medulloblastoma is now divided into four molecular subgroups with distinct characteristics and prognoses. The classification of other unique embryonal tumors is also highlighted. Finally, we present the newest classification of ependymoma; supratentorial ependymomas comprise two subtypes based on expression of the chromosome 11 Open Reading Frame 95-reticuloendotheliosis Viral Oncogene Homolog A or yes-associated protein 1 fusion, whereas posterior fossa ependymomas are divided into two distinct molecular subgroups, posterior fossa-A and posterior fossa-B. SUMMARY: These advances in the molecular classification of pediatric central nervous system tumors have not only assisted in diagnoses, but they have led to a new era of tumor classification and prognostication. They also have served as drivers for the evaluation of new targeted therapies based upon molecular aberrations with the hope for improved survival outcomes for our patients.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Ependymoma , Glioma , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/therapy , Child , Ependymoma/diagnosis , Ependymoma/genetics , Ependymoma/therapy , Glioma/diagnosis , Glioma/genetics , Glioma/therapy , Humans , Mutation
12.
Pediatr Blood Cancer ; 68(8): e28930, 2021 08.
Article in English | MEDLINE | ID: mdl-33565268

ABSTRACT

BACKGROUND: Ependymoma is the third most common malignant CNS tumor in children. Despite multimodal therapy, prognosis of relapsed ependymoma remains poor. Approaches to therapy for relapsed ependymoma are varied. We present a single-institution retrospective review of the outcomes after first relapse of intracranial ependymoma in children. PROCEDURE: We performed a retrospective, IRB-approved chart review of patients with recurrent intracranial ependymoma treated at Dana-Farber/Boston Children's Cancer and Blood Disorders Center from 1990 to 2019. RESULTS: Thirty-four patients with relapsed intracranial ependymoma were identified. At initial diagnosis, 11 patients had supratentorial disease, 22 with posterior fossa disease and one with metastatic disease. Median time-to-first relapse was 14.9 months from initial diagnosis (range 1.4-52.5). Seven patients had metastatic disease at first relapse. Gross total resection (GTR) was associated with improved 5-year progression-free survival (PFS) relative to subtotal resection (STR) and no surgery (p = .005). Localized disease at relapse was associated with improved 5-year overall survival (OS) when compared to metastatic disease (p = .02). Irradiation at first relapse seemed to delay progression but was not associated with statistically prolonged PFS or OS. Tumor location, histology, and chromosomal 1q status did not impact outcome at first relapse, although available molecular data were limited making definitive conclusions difficult. Median time-to-second relapse was 10 months (range 0.7-124). Five-year PFS and OS after first relapse were 19.9% and 45.1%, respectively. Median PFS and OS were 10.0 and 52.5 months after first relapse, respectively. CONCLUSIONS: Relapsed intracranial ependymoma has a poor prognosis despite multimodal therapy. Novel therapeutic strategies are desperately needed for this disease.


Subject(s)
Brain Neoplasms , Ependymoma , Brain Neoplasms/therapy , Child, Preschool , Chronic Disease , Ependymoma/therapy , Humans , Infant , Neoplasm Recurrence, Local/therapy , Retrospective Studies
13.
BMC Biol ; 18(1): 177, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33234154

ABSTRACT

BACKGROUND: Many biological processes, such as cancer metastasis, organismal development, and acquisition of resistance to cytotoxic therapy, rely on the emergence of rare sub-clones from a larger population. Understanding how the genetic and epigenetic features of diverse clones affect clonal fitness provides insight into molecular mechanisms underlying selective processes. While large-scale barcoding with NGS readout has facilitated cellular fitness assessment at the population level, this approach does not support characterization of clones prior to selection. Single-cell genomics methods provide high biological resolution, but are challenging to scale across large populations to probe rare clones and are destructive, limiting further functional analysis of important clones. RESULTS: Here, we develop CloneSifter, a methodology for tracking and enriching rare clones throughout their response to selection. CloneSifter utilizes a CRISPR sgRNA-barcode library that facilitates the isolation of viable cells from specific clones within the barcoded population using a sequence-specific retrieval reporter. We demonstrate that CloneSifter can measure clonal fitness of cancer cell models in vitro and retrieve targeted clones at abundance as low as 1 in 1883 in a heterogeneous cell population. CONCLUSIONS: CloneSifter provides a means to track and access specific and rare clones of interest across dynamic changes in population structure to comprehensively explore the basis of these changes.


Subject(s)
Cloning, Organism/methods , Clustered Regularly Interspaced Short Palindromic Repeats , RNA/metabolism , Cells, Cultured , Clone Cells
14.
J Neurooncol ; 149(2): 253-262, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32780261

ABSTRACT

PURPOSE: Pediatric low-grade gliomas (pLGGs) are the most common CNS tumor of childhood and comprise a heterogenous group of tumors. Children with progressive pLGG often require numerous treatment modalities including surgery, chemotherapy, rarely radiation therapy and, more recently, molecularly targeted therapy. We describe our institutional experience using the MEK inhibitor, trametinib, for recurrent/progressive pLGGs. METHODS: We performed a retrospective, IRB-approved, chart review of all pediatric patients treated with trametinib for recurrent/progressive pLGGs at Dana-Farber/Boston Children's Cancer and Blood Disorder Center between 2016 and 2018. RESULTS: Eleven patients were identified, of which 10 were evaluable for response. Median age at commencement of trametinib treatment was 14.7 years (range 7.3-25.9 years). Tumor molecular status included KIAA1549-BRAF fusion (n = 4), NF1 mutation (n = 4), FGFR mutation (n = 1) and CDKN2A loss (n = 1). Median number of prior treatment regimens was 5 (range 1-12). Median duration of treatment with trametinib was 19.2 months (range 3.8-29.8 months). Based on modified RANO criteria, best responses included partial (n = 2), minor response (n = 2) and stable disease (n = 6). Two patients remain on therapy (29.8 and 25.9 months, respectively). The most common toxicities attributable to trametinib were rash, fatigue and gastrointestinal disturbance. Five patients required dose reduction for toxicities. Two patients experienced significant intracranial hemorrhage (ICH) while on trametinib. While it is unclear whether ICH was directly attributable to trametinib, therapy was discontinued. CONCLUSION: Trametinib appears to be an effective treatment for patients with recurrent/progressive pLGG. The toxicities of this therapy warrant further investigation, with particular attention to the potential risk for intracranial hemorrhage. Early phase multi-institutional clinical trials are underway.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Adolescent , Adult , Brain Neoplasms/pathology , Child , Female , Follow-Up Studies , Glioma/pathology , Humans , Male , Neoplasm Recurrence, Local/pathology , Prognosis , Retrospective Studies , Young Adult
15.
J Neurooncol ; 145(2): 349-355, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31571114

ABSTRACT

BACKGROUND: Pediatric brain tumors are associated with high morbidity and mortality, in part due to insufficient understanding of tumor biology. With limited tissue allocation for research from surgical specimens, a key barrier to improving biological understanding, brain tumor autopsies have become an increasingly valuable resource. This study reviews the brain tumor autopsy practice at our institution and describes specific emerging research utilization patterns beyond the clinical autopsy report. METHODS: We performed a retrospective analysis of brain tumor autopsies at Boston Children's Hospital (BCH) between 2007 and 2017 and reviewed their consents, neuropathology reports and final diagnoses. We reviewed the method of tissue triaging for research consented autopsies (bioregistry, frozen and fresh tissue) and documented their specific uses. RESULTS: Ninety-six deaths at BCH were due to brain tumors; 56 autopsies were performed (58.3%), of which 49 (87.5%) were consented for research. Tumor mapping was performed on all cases and tissue was allocated for DNA- and RNA-based sequencing studies (published and ongoing). Three tissue allocations with a postmortem interval of 8 h or less resulted in successful cell lines. Tissue from 14 autopsies was contributed to the National DIPG Registry. CONCLUSION: Our institutional pediatric brain tumor autopsy clinical experience demonstrates the increased utility and wide utilization of autopsy-derived tissue for multiple types of research. These results support the increased efforts to obtain research consent for brain tumor autopsy and active collection of unfixed autopsy material in the molecular era.


Subject(s)
Autopsy/methods , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Medical Oncology/methods , Biomedical Research , Child , Humans , Retrospective Studies
16.
Curr Opin Pediatr ; 31(1): 21-27, 2019 02.
Article in English | MEDLINE | ID: mdl-30531227

ABSTRACT

PURPOSE OF REVIEW: Pediatric low-grade gliomas (pLGGs) have been treated with similar therapies for the last 30 years. Recent biological insights have allowed a new generation of targeted therapies to be developed for these diverse tumors. At the same time, technological advances may redefine the late toxicities associated with radiation therapy. Understanding recent developments in pLGG therapy is essential to the management of these common pediatric tumors. RECENT FINDINGS: It is now well understood that aberrations of the mitogen-activated protein kinase pathway are key to oncogenesis in low-grade gliomas. This understanding, along with the development of available targeted agents, have heralded a new era of understanding and treatment for these patients. Promising, sustained responses are now being seen in early phase trials among patients with multiply recurrent/progressive disease. Also, newer and highly conformal radiation approaches such as proton beam radiotherapy maintain efficacy of radiation but limit radiation-associated toxicities. SUMMARY: Novel therapies offer the potential for tumor control with greatly reduced toxicities. However, late effects of these therapies are just now being explored. Improved radiation approaches and targeted agents have the potential to redefine traditional therapy for pLGG.


Subject(s)
Glioma/therapy , Child , Glioma/pathology , Humans , Neoplasm Grading
17.
Nature ; 555(7696): 316-317, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29542703

Subject(s)
Neoplasms , Humans
18.
J Neurooncol ; 137(2): 269-278, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29322427

ABSTRACT

There is little known regarding the immune infiltrate present in pediatric brain tumors and how this compares to what is known about histologically similar adult tumors and its correlation with survival. Here, we provide a descriptive analysis of the immune infiltrate of 22 fresh pediatric brain tumor tissue samples of mixed diagnoses and 40 peripheral blood samples. Samples were analyzed using a flow cytometry panel containing markers for immune cell subtypes, costimulatory markers, inhibitory signals, and markers of activation. This was compared to the standard method of immunohistochemistry (IHC) for immune markers for 89 primary pediatric brain tumors. Both flow cytometry and IHC data did not correlate with the grade of tumor or mutational load and IHC data was not significantly associated with survival for either low grade or high grade gliomas. There is a trend towards a more immunosuppressive phenotype in higher grade tumors with more regulatory T cells present in these tumor types. Both PD1 and PDL1 were present in only a small percentage of the tumor infiltrate. T cell receptor sequencing revealed up to 10% clonality of T cells in tumor infiltrates and no significant difference in clonality between low and high grade gliomas. We have shown the immune infiltrate of pediatric brain tumors does not appear to correlate with grade or survival for a small sample of patients. Further research and larger studies are needed to fully understand the interaction of pediatric brain tumors and the immune system.


Subject(s)
Brain Neoplasms/immunology , Adolescent , B7-H1 Antigen/metabolism , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Brain/immunology , Brain/pathology , Brain Neoplasms/blood , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Child, Preschool , Humans , Immunophenotyping , Infant , Mutation , Neoplasm Grading , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/pathology
19.
Nature ; 550(7676): 333, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29052624

Subject(s)
Publications , Publishing , Humans
20.
Proc Natl Acad Sci U S A ; 112(1): 232-7, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535366

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) is the most common and aggressive form of epithelial ovarian cancer, for which few targeted therapies exist. To search for new therapeutic target proteins, we performed an in vivo shRNA screen using an established human HGSOC cell line growing either subcutaneously or intraperitoneally in immunocompromised mice. We identified genes previously implicated in ovarian cancer such as AURKA1, ERBB3, CDK2, and mTOR, as well as several novel candidates including BRD4, VRK1, and GALK2. We confirmed, using both genetic and pharmacologic approaches, that the activity of BRD4, an epigenetic transcription modulator, is necessary for proliferation/survival of both an established human ovarian cancer cell line (OVCAR8) and a subset of primary serous ovarian cancer cell strains (DFs). Among the DFs tested, the strains sensitive to BRD4 inhibition revealed elevated expression of either MYCN or c-MYC, with MYCN expression correlating closely with JQ1 sensitivity. Accordingly, primary human xenografts derived from high-MYCN or c-MYC strains exhibited sensitivity to BRD4 inhibition. These data suggest that BRD4 inhibition represents a new therapeutic approach for MYC-overexpressing HGSOCs.


Subject(s)
Genetic Testing , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/therapy , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Transcription Factors/metabolism , Animals , Carcinoma, Ovarian Epithelial , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Female , Genetic Association Studies , Humans , Mice , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL