Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Neuro Oncol ; 21(12): 1540-1551, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31276586

ABSTRACT

BACKGROUND: Treatment for pediatric posterior fossa group A (PFA) ependymoma with gain of chromosome 1q (1q+) has not improved over the past decade owing partially to lack of clinically relevant models. We described the first 2 1q+ PFA cell lines, which have significantly enhanced our understanding of PFA tumor biology and provided a tool to identify specific 1q+ PFA therapies. However, cell lines do not accurately replicate the tumor microenvironment. Our present goal is to establish patient-derived xenograft (PDX) mouse models. METHODS: Disaggregated tumors from 2 1q+ PFA patients were injected into the flanks of NSG mice. Flank tumors were then transplanted into the fourth ventricle or lateral ventricle of NSG mice. Characterization of intracranial tumors was performed using imaging, histology, and bioinformatics. RESULTS: MAF-811_XC and MAF-928_XC established intracranially within the fourth ventricle and retained histological, methylomic, and transcriptomic features of primary patient tumors. We tested the feasibility of treating PDX mice with fractionated radiation or chemotherapy. Mice tolerated radiation despite significant tumor burden, and follow-up imaging confirmed radiation can reduce tumor size. Treatment with fluorouracil reduced tumor size but did not appear to prolong survival. CONCLUSIONS: MAF-811_XC and MAF-928_XC are novel, authentic, and reliable models for studying 1q+ PFA in vivo. Given the successful response to radiation, these models will be advantageous for testing clinically relevant combination therapies to develop future clinical trials for this high-risk subgroup of pediatric ependymoma.


Subject(s)
Brain Neoplasms/pathology , Chemoradiotherapy/mortality , Chromosomes, Human, Pair 1/genetics , Disease Models, Animal , Ependymoma/pathology , Infratentorial Neoplasms/pathology , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Proliferation , Child , Ependymoma/genetics , Ependymoma/therapy , Humans , Infratentorial Neoplasms/genetics , Infratentorial Neoplasms/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 17(9): 1984-1994, 2018 09.
Article in English | MEDLINE | ID: mdl-29925527

ABSTRACT

Children with ependymoma (EPN) are cured in less than 50% of cases, with little improvement in outcome over the last several decades. Chemotherapy has not affected survival in EPN, due in part to a lack of preclinical models that has precluded comprehensive drug testing. We recently developed two human EPN cell lines harboring high-risk phenotypes which provided us with an opportunity to execute translational studies. EPN and other pediatric brain tumor cell lines were subject to a large-scale comparative drug screen of FDA-approved oncology drugs for rapid clinical application. The results of this in vitro study were combined with in silico prediction of drug sensitivity to identify EPN-selective compounds, which were validated by dose curve and time course modeling. Mechanisms of EPN-selective antitumor effect were further investigated using transcriptome and proteome analyses. We identified three classes of oncology drugs that showed EPN-selective antitumor effect, namely, (i) fluorinated pyrimidines (5-fluorouracil, carmofur, and floxuridine), (ii) retinoids (bexarotene, tretinoin and isotretinoin), and (iii) a subset of small-molecule multireceptor tyrosine kinase inhibitors (axitinib, imatinib, and pazopanib). Axitinib's antitumor mechanism in EPN cell lines involved inhibition of PDGFRα and PDGFRß and was associated with reduced mitosis-related gene expression and cellular senescence. The clinically available, EPN-selective oncology drugs identified by our study have the potential to critically inform design of upcoming clinical studies in EPN, in particular for those children with recurrent EPN who are in the greatest need of novel therapeutic approaches. Mol Cancer Ther; 17(9); 1984-94. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Drug Screening Assays, Antitumor/methods , Ependymoma/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Child , Computer Simulation , Drug Approval , Ependymoma/drug therapy , Ependymoma/pathology , Humans , Risk Factors
3.
Pediatr Blood Cancer ; 65(5): e26960, 2018 05.
Article in English | MEDLINE | ID: mdl-29350470

ABSTRACT

BACKGROUND: A desperate need for novel therapies in pediatric ependymoma (EPN) exists, as chemotherapy remains ineffective and radiotherapy often fails. EPN have significant infiltration of immune cells, which correlates with outcome. Immune checkpoint inhibitors provide an avenue for new treatments. This study characterizes tumor-infiltrating immune cells in EPN and aims at predicting candidates for clinical trials using checkpoint inhibitors targeting PD-L1/PD-1 (programmed death ligand 1/programmed death 1). METHODS: The transcriptomic profiles of the primary study cohort of EPN and other pediatric brain tumors were interrogated to identify PD-L1 expression levels. Transcriptomic findings were validated using the western blotting, immunohistochemistry and flow cytometry. RESULTS: We evaluated PD-L1 mRNA expression across four intracranial subtypes of EPN in two independent cohorts and found supratentorial RELA fusion (ST-RELA) tumors to have significantly higher levels. There was a correlation between high gene expression and protein PD-L1 levels in ST-RELA tumors by both the western blot and immunohistochemisty. The investigation of EPN cell populations revealed PD-L1 was expressed on both tumor and myeloid cells in ST-RELA. Other subtypes had little PD-L1 in either tumor or myeloid cell compartments. Lastly, we measured PD-1 levels on tumor-infiltrating T cells and found ST-RELA tumors express PD-1 in both CD4 and CD8 T cells. A functional T-cell exhaustion assay found ST-RELA T cells to be exhausted and unable to secrete IFNγ on stimulation. CONCLUSIONS: These findings in ST-RELA suggest tumor evasion and immunsuppression due to PD-L1/PD-1-mediated T-cell exhaustion. Trials of checkpoint inhibitors in EPN should be enriched for ST-RELA tumors.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Ependymoma/metabolism , Supratentorial Neoplasms/metabolism , Transcription Factor RelA/metabolism , Adolescent , Adult , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Child , Child, Preschool , Cohort Studies , Ependymoma/genetics , Ependymoma/pathology , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Infant , Male , Molecular Targeted Therapy , Prognosis , Supratentorial Neoplasms/genetics , Supratentorial Neoplasms/pathology , T-Lymphocytes/metabolism , Transcription Factor RelA/genetics , Young Adult
4.
J Neuropathol Exp Neurol ; 76(9): 779-788, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28859336

ABSTRACT

Pediatric adamantinomatous craniopharyngioma (ACP) is a highly solid and cystic tumor, often causing substantial damage to critical neuroendocrine structures such as the hypothalamus, pituitary gland, and optic apparatus. Paracrine signaling mechanisms driving tumor behavior have been hypothesized, with IL-6R overexpression identified as a potential therapeutic target. To identify potential novel therapies, we characterized inflammatory and immunomodulatory factors in ACP cyst fluid and solid tumor components. Cytometric bead analysis revealed a highly pro-inflammatory cytokine pattern in fluid from ACP compared to fluids from another cystic pediatric brain tumor, pilocytic astrocytoma. Cytokines and chemokines with particularly elevated concentrations in ACPs were IL-6, CXCL1 (GRO), CXCL8 (IL-8) and the immunosuppressive cytokine IL-10. These data were concordant with solid tumor compartment transcriptomic data from a larger cohort of ACPs, other pediatric brain tumors and normal brain. The majority of receptors for these cytokines and chemokines were also over-expressed in ACPs. In addition to IL-10, the established immunosuppressive factor IDO-1 was overexpressed by ACPs at the mRNA and protein levels. These data indicate that ACP cyst fluids and solid tumor components are characterized by an inflammatory cytokine and chemokine expression pattern. Further study regarding selective cytokine blockade may inform novel therapeutic interventions.


Subject(s)
Craniopharyngioma/metabolism , Cyst Fluid/metabolism , Cytokines/metabolism , Pituitary Neoplasms/metabolism , Child , Child, Preschool , Cohort Studies , Craniopharyngioma/genetics , Craniopharyngioma/pathology , Cyst Fluid/immunology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Microarray Analysis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , RNA, Messenger/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism
5.
J Neuropathol Exp Neurol ; 76(7): 595-604, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28863455

ABSTRACT

Ependymoma (EPN) is a common brain tumor of childhood that, despite standard surgery and radiation therapy, has a relapse rate of 50%. Clinical trials have been unsuccessful in improving outcome by addition of chemotherapy, and identification of novel therapeutics has been hampered by a lack of in vitro and in vivo models. We describe 2 unique EPN cell lines (811 and 928) derived from recurrent intracranial metastases. Both cell lines harbor the high-risk chromosome 1q gain (1q+) and a derivative chromosome 6, and both are classified as molecular group A according to transcriptomic analysis. Transcriptional enrichment of extracellular matrix-related genes was a common signature of corresponding primary tumors and cell lines in both monolayer and 3D formats. EPN cell lines, when cultured in 3D format, clustered closer to the primary tumors with better fidelity of EPN-specific transcripts than when grown as a monolayer. Additionally, 3D culture revealed ependymal rosette formation and cilia-related ontologies, similar to in situ tumors. Our data confirm the validity of the 811 and 928 cell lines as representative models of intracranial, posterior fossa 1q+ EPN, which holds potential to advance translational science for patients affected by this tumor.


Subject(s)
Cell Line, Tumor/pathology , Chromosome Aberrations , Chromosomes, Human, Pair 1/genetics , Ependymoma/pathology , Infratentorial Neoplasms/genetics , Infratentorial Neoplasms/pathology , Child , Cytogenetic Analysis , DNA-Binding Proteins/metabolism , Ependymoma/genetics , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Humans , Imaging, Three-Dimensional , Ki-67 Antigen/metabolism , Male , Microarray Analysis , Microscopy, Confocal , Mucin-1/metabolism , Nuclear Proteins/metabolism , Receptors, HIV/metabolism , Transcription Factors/metabolism
6.
Neuro Oncol ; 19(10): 1350-1360, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28510691

ABSTRACT

BACKGROUND: Inflammation has been identified as a hallmark of high-risk Group A (GpA) ependymoma (EPN). Chronic interleukin (IL)-6 secretion from GpA tumors drives an immune suppressive phenotype by polarizing infiltrating monocytes. This study determines the mechanism by which IL-6 is dysregulated in GpA EPN. METHODS: Twenty pediatric GpA and 21 pediatric Group B (GpB) EPN had gene set enrichment analysis for MSigDB Hallmark gene sets performed. Protein and RNA from patients and cell lines were used to validate transcriptomic findings. GpA cell lines 811 and 928 were used for in vitro experiments performed in this study. RESULTS: The nuclear factor-kappaB (NF-κB) pathway is a master regulator of IL-6 and a signaling pathway enriched in GpA compared with GpB EPN. Knockdown of NF-κB led to significant downregulation of IL-6 in 811 and 928. NF-κB activation was independent of tumor necrosis factor alpha (TNF-α) stimulation in both cell lines, suggesting that NF-κB hyperactivation is mediated through an alternative mechanism. Leucine zipper downregulated in cancer 1 (LDOC1) is a known transcriptional repressor of NF-κB. In many cancers, LDOC1 promoter is methylated, which inhibits gene transcription. We found decreased LDOC1 gene expression in GpA compared with GpB EPN, and in other pediatric brain tumors. EPN cells treated with 5AZA-DC, demethylated LDOC1 regulatory regions, upregulated LDOC1 expression, and concomitantly decreased IL-6 secretion. Stable knockdown of LDOC1 in EPN cell lines resulted in a significant increase in gene transcription of v-rel avian reticuloendotheliosis viral oncogene homolog A, which correlated to an increase in NF-κB target genes. CONCLUSION: These results suggest that epigenetic silencing of LDOC1 in GpA EPN regulates tumor biology and drives inflammatory immune phenotype.


Subject(s)
Ependymoma/metabolism , Immunophenotyping , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Ependymoma/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunophenotyping/methods , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Transcriptional Activation/physiology , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...