Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Clin Cancer Res ; 30(11): 2545-2557, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38551501

ABSTRACT

PURPOSE: Medulloblastoma (MB), the most common childhood malignant brain tumor, has a poor prognosis in about 30% of patients. The current standard of care, which includes surgery, radiation, and chemotherapy, is often responsible for cognitive, neurologic, and endocrine side effects. We investigated whether chimeric antigen receptor (CAR) T cells directed toward the disialoganglioside GD2 can represent a potentially more effective treatment with reduced long-term side effects. EXPERIMENTAL DESIGN: GD2 expression was evaluated on primary tumor biopsies of MB children by flow cytometry. GD2 expression in MB cells was also evaluated in response to an EZH2 inhibitor (tazemetostat). In in vitro and in vivo models, GD2+ MB cells were targeted by a CAR-GD2.CD28.4-1BBζ (CAR.GD2)-T construct, including the suicide gene inducible caspase-9. RESULTS: GD2 was expressed in 82.68% of MB tumors. The SHH and G3-G4 subtypes expressed the highest levels of GD2, whereas the WNT subtype expressed the lowest. In in vitro coculture assays, CAR.GD2 T cells were able to kill GD2+ MB cells. Pretreatment with tazemetostat upregulated GD2 expression, sensitizing GD2dimMB cells to CAR.GD2 T cells cytotoxic activity. In orthotopic mouse models of MB, intravenously injected CAR.GD2 T cells significantly controlled tumor growth, prolonging the overall survival of treated mice. Moreover, the dimerizing drug AP1903 was able to cross the murine blood-brain barrier and to eliminate both blood-circulating and tumor-infiltrating CAR.GD2 T cells. CONCLUSIONS: Our experimental data indicate the potential efficacy of CAR.GD2 T-cell therapy. A phase I/II clinical trial is ongoing in our center (NCT05298995) to evaluate the safety and therapeutic efficacy of CAR.GD2 therapy in high-risk MB patients.


Subject(s)
Gangliosides , Immunotherapy, Adoptive , Medulloblastoma , Receptors, Chimeric Antigen , Xenograft Model Antitumor Assays , Humans , Medulloblastoma/therapy , Medulloblastoma/immunology , Medulloblastoma/pathology , Medulloblastoma/genetics , Medulloblastoma/metabolism , Animals , Mice , Gangliosides/metabolism , Gangliosides/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Cell Line, Tumor , Child , Female , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cerebellar Neoplasms/therapy , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/metabolism , Morpholines/pharmacology , Male , Child, Preschool , Benzamides , Biphenyl Compounds , Pyridones
2.
Front Immunol ; 13: 911260, 2022.
Article in English | MEDLINE | ID: mdl-35967388

ABSTRACT

Medulloblastoma, a common pediatric malignant tumor, has been recognized to have four molecular subgroups [wingless (WNT), sonic hedgehog (SHH), group 3, group 4], which are defined by the characteristic gene transcriptomic and DNA methylomic profiles, and has distinct clinical features within each subgroup. The tumor immune microenvironment is integral in tumor initiation and progression and might be associated with therapeutic responses. However, to date, the immune infiltrative landscape of medulloblastoma has not yet been elucidated. Thus, we proposed MethylCIBERSORT to estimate the degree of immune cell infiltration and weighted correlation network analysis (WGCNA) to find modules of highly correlated genes. Synthesizing the hub genes in the protein-protein interaction (PPI) network and modules of the co-expression network, we identify three candidate biomarkers [GRB2-associated-binding protein 1 (GAB1), Abelson 1 (ABL1), and CXC motif chemokine receptor type 4 (CXCR4)] via the molecular profiles of medulloblastoma. Given this, we investigated the correlation between these three immune hub genes and immune checkpoint blockade response and the potential of drug prediction further. In addition, this study demonstrated a higher presence of endothelial cells and infiltrating immune cells in Group 3 tumor bulk. The above results will be conducive to better comprehending the immune-related pathogenesis and treatment of medulloblastoma.


Subject(s)
Adaptor Proteins, Signal Transducing , Cerebellar Neoplasms , Medulloblastoma , Proto-Oncogene Proteins c-abl , Receptors, CXCR4 , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Biomarkers , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Child , Endothelial Cells/immunology , Hedgehog Proteins/immunology , Humans , Medulloblastoma/genetics , Medulloblastoma/immunology , Medulloblastoma/pathology , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
3.
Hum Mol Genet ; 30(18): 1721-1733, 2021 08 28.
Article in English | MEDLINE | ID: mdl-33949667

ABSTRACT

Medulloblastoma, a common pediatric malignant brain tumor, consists of four distinct molecular subgroups WNT, SHH, Group 3 and Group 4. Exome sequencing of 11 WNT subgroup medulloblastomas from an Indian cohort identified mutations in several chromatin modifier genes, including genes of the mammalian SWI/SNF complex. The genome of WNT subgroup tumors is known to be stable except for monosomy 6. Two tumors, having monosomy 6, carried a loss of function mutation in the ARID1B gene located on chromosome 6. ARID1B expression is also lower in the WNT subgroup tumors compared to other subgroups and normal cerebellar tissues that could result in haploinsufficiency. The short hairpin RNA-mediated knockdown of ARID1B expression resulted in a significant increase in the malignant potential of medulloblastoma cells. Transcriptome sequencing identified upregulation of several genes encoding cell adhesion proteins, matrix metalloproteases indicating the epithelial-mesenchymal transition. The ARID1B knockdown also upregulated ERK1/ERK2 and PI3K/AKT signaling with a decrease in the expression of several negative regulators of these pathways. The expression of negative regulators of the WNT signaling like TLE1, MDFI, GPX3, ALX4, DLC1, MEST decreased upon ARID1B knockdown resulting in the activation of the canonical WNT signaling pathway. Synthetic lethality has been reported between SWI/SNF complex mutations and EZH2 inhibition, suggesting EZH2 inhibition as a possible therapeutic modality for WNT subgroup medulloblastomas. Thus, the identification of ARID1B as a tumor suppressor and its downregulation resulting in the activation of multiple signaling pathways opens up opportunities for novel therapeutic modalities for the treatment of WNT subgroup medulloblastoma.


Subject(s)
Cerebellar Neoplasms/metabolism , DNA-Binding Proteins/biosynthesis , Down-Regulation , Gene Expression Regulation, Neoplastic , Medulloblastoma/metabolism , Transcription Factors/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Child , DNA-Binding Proteins/genetics , Female , Humans , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism
4.
Int J Radiat Oncol Biol Phys ; 111(2): 479-490, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33974888

ABSTRACT

PURPOSE: Radiation therapy (RT) modulates immune cells and cytokines, resulting in both clinically beneficial and detrimental effects. The changes in peripheral blood T lymphocyte subsets and cytokines during RT for pediatric brain tumors and the association of these changes with therapeutic outcomes have not been well described. METHODS AND MATERIALS: The study population consisted of children (n = 83, aged 3~18) with primary brain tumors (medulloblastoma, glioma, germ cell tumors (GCT), and central nervous system embryonal tumor-not otherwise specified), with or without residual or disseminated (R/D) diseases who were starting standard postoperative focal or craniospinal irradiation (CSI). Peripheral blood T lymphocyte subsets collected before and 4 weeks after RT were enumerated by flow cytometry. Plasma levels of interleukin (IL)-2, IL-4, IL-6, IL-10, tumor necrosis factor-α, interferon-γ, and IL-17A were measured by cytometric bead array. RESULTS: Patients with R/D lesions receiving CSI (n = 32) had a post-RT increase in the frequency of CD3+T and CD8+T cells, a decrease in CD4+T cells, and an increase in regulatory T cells (Tregs) and CD8+CD28- suppressor cells, which was more predominantly seen in these patients than in other groups. In the CSI group with such R/D lesions, consisting of patients with medulloblastoma and germ cell tumors, 19 experienced a complete response (CR) and 13 experienced a partial response (PR) on imaging at 4 weeks after RT. The post/pre-RT ratio of Tregs (P = .0493), IL-6 (P = .0111), and IL-10 (P = .0070) was lower in the CR group than in the PR group. Multivariate analysis revealed that the post/pre-RT ratios of Treg, IL-6, and IL-10 were independent predictors of CR (P < .0001, P = .018, P < .0001, respectively). The areas under the receiver operating curves and confidence intervals were 0.7652 (0.5831-0.8964), 0.7794 (0.5980-0.9067), and 0.7085 (0.5223-0.8552) for IL-6, IL-10, and Treg, respectively. The sensitivities of IL-6, IL-10, and Treg to predict radiotherapeutic responses were 100%, 92.3%, and 61.5%, and specificity was 52.6%, 57.9%, and 84.2%, respectively. CONCLUSIONS: CSI treatment to those with R/D lesions predominantly exerted an effect on antitumor immune response compared with both R/D lesion-free but exposed to focal or CSI RT and with R/D lesions and exposed to focal RT. Such CSI with R/D lesions group experiencing CR is more likely to have a decrease in immunoinhibitory molecules and cells than patients who only achieve PR. Measuring peripheral blood Treg, IL-6, and IL-10 levels could be valuable for predicting radiotherapeutic responses of pediatric brain tumors with R/D lesions to CSI for medulloblastoma and intracranial germ cell tumors.


Subject(s)
Cerebellar Neoplasms/radiotherapy , Craniospinal Irradiation , Interleukin-10/blood , Interleukin-6/blood , Medulloblastoma/radiotherapy , Neoplasms, Germ Cell and Embryonal/radiotherapy , T-Lymphocytes, Regulatory/immunology , Adolescent , Cerebellar Neoplasms/immunology , Child , Child, Preschool , Female , Humans , Logistic Models , Male , Medulloblastoma/immunology , Medulloblastoma/pathology , Neoplasms, Germ Cell and Embryonal/immunology , Neoplasms, Germ Cell and Embryonal/pathology , T-Lymphocyte Subsets/immunology
5.
Cell Rep ; 34(13): 108917, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789113

ABSTRACT

Tumor-associated macrophages (TAMs) play an important role in tumor immunity and comprise of subsets that have distinct phenotype, function, and ontology. Transcriptomic analyses of human medulloblastoma, the most common malignant pediatric brain cancer, showed that medulloblastomas (MBs) with activated sonic hedgehog signaling (SHH-MB) have significantly more TAMs than other MB subtypes. Therefore, we examined MB-associated TAMs by single-cell RNA sequencing of autochthonous murine SHH-MB at steady state and under two distinct treatment modalities: molecular-targeted inhibitor and radiation. Our analyses reveal significant TAM heterogeneity, identify markers of ontologically distinct TAM subsets, and show the impact of brain microenvironment on the differentiation of tumor-infiltrating monocytes. TAM composition undergoes dramatic changes with treatment and differs significantly between molecular-targeted and radiation therapy. We identify an immunosuppressive monocyte-derived TAM subset that emerges with radiation therapy and demonstrate its role in regulating T cell and neutrophil infiltration in MB.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/therapy , Hedgehog Proteins/metabolism , Macrophages/metabolism , Macrophages/pathology , Medulloblastoma/pathology , Medulloblastoma/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/immunology , Genetic Markers , Humans , Medulloblastoma/genetics , Medulloblastoma/immunology , Mice , Microglia/pathology , Monocytes/pathology , Single-Cell Analysis , Transcription, Genetic , Tumor Microenvironment
6.
Biosci Rep ; 41(1)2021 01 29.
Article in English | MEDLINE | ID: mdl-33345275

ABSTRACT

Medulloblastoma (MB) is one of the most common central nervous system tumors in children. At present, the vital role of immune abnormalities has been proved in tumorigenesis and progression. However, the immune mechanism in MB is still poorly understood. In the present study, 51 differentially expressed immune-related genes (DE-IRGs) and 226 survival associated immune-related genes (Sur-IRGs) were screened by an integrated analysis of multi-array. Moreover, the potential pathways were enriched by functional analysis, such as 'cytokine-cytokine receptor interaction', 'Ras signaling pathway', 'PI3K-Akt signaling pathway' and 'pathways in cancer'. Furthermore, 10 core IRGs were identified from DE-IRGs and Sur-IRGs. And the potential regulatory mechanisms of core IRGs were also explored. Additionally, a new prognostic model, including 7 genes (HDGF, CSK, PNOC, S100A13, RORB, FPR1, and ICAM2) based on IRGs, was established by multivariable COX analysis. In summary, our study revealed the underlying immune mechanism of MB. Moreover, we developed a prognostic model associated with clinical characteristics and could reflect the infiltration of immune cells.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/immunology , Medulloblastoma/genetics , Medulloblastoma/immunology , Models, Biological , Cerebellar Neoplasms/pathology , Gene Regulatory Networks , Humans , Medulloblastoma/pathology , Prognosis , Transcription Factors/genetics
7.
Cancer Lett ; 499: 188-200, 2021 02 28.
Article in English | MEDLINE | ID: mdl-33253789

ABSTRACT

The deregulation of epigenetic pathways has been implicated as a critical step in tumorigenesis including in childhood brain tumor medulloblastoma. The H3K27me3 demethylase UTX/KDM6A plays important roles in development and is frequently mutated in various types of cancer. However, how UTX regulates tumor development remains largely unclear. Here, we report the generation of a UTX-deleted mouse model of SHH medulloblastoma that demonstrates the tumor suppressor functions of UTX, which could be antagonized by the deletion of another H3K27me3 demethylase JMJD3/KDM6B. Intriguingly, UTX deletion in cancerous cerebellar granule neuron precursors (CGNPs) resulted in the impaired recruitment of host CD8+ T cells to the tumor microenvironment through a non-cell autonomous mechanism. In both mouse medulloblastoma models and in human medulloblastoma cells, we showed that UTX activates Th1-type chemokines, which are responsible for T cell migration. Surprisingly, our results showed that the depletion of cytotoxic CD8+ T cells did not affect mouse medulloblastoma growth. Nevertheless, the UTX/chemokine/T cell recruitment pathway we identified may be applied to many other cancers and may be important for improving cancer immunotherapy. In addition, UTX is required for the expression of NeuroD2 in precancerous progenitors, which encodes a potent proneural transcription factor. Overexpression of NEUROD2 in CGNPs decreased cell proliferation and increased neuron differentiation. We showed that UTX deletion led to impaired neural differentiation, which could coordinate with active SHH signaling to accelerate medulloblastoma development. Thus, UTX regulates both cell-intrinsic oncogenic processes and the tumor microenvironment in medulloblastoma. Our study provides insights into both medulloblastoma development and context dependent functions of UTX in tumorigenesis.


Subject(s)
Cerebellar Neoplasms/genetics , Histone Demethylases/metabolism , Medulloblastoma/genetics , Tumor Suppressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Cerebellum/cytology , Cerebellum/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/immunology , Gene Knockdown Techniques , Histone Demethylases/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Male , Medulloblastoma/immunology , Medulloblastoma/pathology , Mice , Mice, Transgenic , Neoplastic Stem Cells/pathology , Neurons/pathology , Neuropeptides/genetics , Primary Cell Culture , Smoothened Receptor/genetics , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Suppressor Proteins/genetics
8.
Hum Gene Ther ; 31(19-20): 1132-1139, 2020 10.
Article in English | MEDLINE | ID: mdl-32657154

ABSTRACT

Brain tumors represent the most common pediatric solid neoplasms and leading cause of childhood cancer-related morbidity and mortality. Although most adult brain tumors are supratentorial and arise in the cerebrum, the majority of pediatric brain tumors are infratentorial and arise in the posterior fossa, specifically the cerebellum. Outcomes from malignant cerebellar tumors are unacceptable despite aggressive treatments (surgery, radiation, and/or chemotherapy) that are harmful to the developing brain. Novel treatments/approaches such as oncolytic virotherapy are urgently needed. Preclinical and prior clinical studies suggest that genetically engineered oncolytic herpes simplex virus (HSV-1) G207 can safely target cerebellar malignancies and has potential to induce an antitumor immune response at local and distant sites of disease, including spinal metastases and leptomeningeal disease. Herein, we outline the rationale, design, and significance of a first-in-human immunotherapy Phase 1 clinical trial targeting recurrent cerebellar malignancies with HSV G207 combined with a single low-dose of radiation (5 Gy), designed to enhance virus replication and innate and adaptive immune responses. We discuss the unique challenges of inoculating virus through intratumoral catheters into cerebellar tumors. The trial utilizes a single arm open-label traditional 3 + 3 design with four dose cohorts. The primary objective is to assess safety and tolerability of G207 with radiation in recurrent/progressive malignant pediatric cerebellar tumors. After biopsy to prove recurrence/progression, one to four intratumoral catheters will be placed followed by a controlled-rate infusion of G207 for 6 h followed by the removal of catheters at the bedside. Radiation will be given within 24 h of virus inoculation. Patients will be monitored closely for toxicity and virus shedding. Efficacy will be assessed by measuring radiographic response, performance score, progression-free and overall survival, and quality of life. The data obtained will be invaluable in our efforts to produce more effective and less toxic therapies for children with high-grade brain tumors.


Subject(s)
Cerebellar Neoplasms/therapy , Oncolytic Virotherapy/methods , Radiotherapy/methods , Simplexvirus/genetics , Adolescent , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Clinical Trials, Phase I as Topic , Cohort Studies , Combined Modality Therapy , Female , Humans , Male , Virus Replication
9.
BMC Cancer ; 20(1): 554, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32539808

ABSTRACT

BACKGROUND: Metastatic medulloblastoma (MB) portends a poor prognosis. Amongst the 4 molecular subtypes, Group 3 and Group 4 patients have a higher incidence of metastatic disease, especially involving the neuroaxis. At present, mechanisms underlying MB metastasis remain elusive. Separately, inflammation has been implicated as a key player in tumour development and metastasis. Cytokines and their inflammation-related partners have been demonstrated to act on autocrine and, or paracrine pathways within the tumour microenvironment for various cancers. In this study, the authors explore the involvement of cerebrospinal fluid (CSF) cytokines in Group 3 and 4 MB patients with disseminated disease. METHODS: This is an ethics approved, retrospective study of prospectively collected data based at a single institution. Patient clinicpathological data and corresponding bio-materials are collected after informed consent. All CSF samples are interrogated using a proteomic array. Resultant expression data of selected cytokines are correlated with each individual's clinical information. Statistical analysis is employed to determine the significance of the expression of CSF cytokines in Group 3 and 4 patients with metastatic MB versus non-metastatic MB. RESULTS: A total of 10 patients are recruited for this study. Median age of the cohort is 6.6 years old. Based on Nanostring gene expression analysis, 5 patients have Group 3 as their molecular subtype and the remaining 5 are Group 4. There are 2 non-metastatic versus 3 metastatic patients within each molecular subtype. Proteomic CSF analysis of all patients for both subtypes show higher expression of CCL2 in the metastatic group versus the non-metastatic group. Within the Group 3 subtype, the MYC-amplified Group 3 MB patients with existing and delayed metastases express higher levels of CXCL1, IL6 and IL8 in their CSF specimens at initial presentation. Furthermore, a longitudinal study of metastatic Group 3 MB observes that selected cytokines are differentially expressed in MYC-amplified metastatic Group 3 MB, in comparison to the non-MYC amplified metastatic Group 3 MB patient. CONCLUSION: This study demonstrates higher expression of selected CSF cytokines, in particular CCL2, in metastatic Group 3 and 4 MB patients. Although our results are preliminary, they establish a proof-of-concept basis for continued work in a larger cohort of patients affected by this devastating disease.


Subject(s)
Biomarkers, Tumor/cerebrospinal fluid , Cerebellar Neoplasms/pathology , Cytokines/cerebrospinal fluid , Medulloblastoma/diagnosis , Biomarkers, Tumor/immunology , Brain/diagnostic imaging , Cerebellar Neoplasms/cerebrospinal fluid , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/surgery , Child , Child, Preschool , Cytokines/immunology , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Male , Medulloblastoma/cerebrospinal fluid , Medulloblastoma/secondary , Medulloblastoma/surgery , Proof of Concept Study , Prospective Studies , Proteomics , Retrospective Studies
10.
Int J Mol Sci ; 21(12)2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32585856

ABSTRACT

Medulloblastomas arise from undifferentiated precursor cells in the cerebellum and account for about 20% of all solid brain tumors during childhood; standard therapies include radiation and chemotherapy, which oftentimes come with severe impairment of the cognitive development of the young patients. Here, we show that the posttranscriptional regulator Y-box binding protein 1 (YBX1), a DNA- and RNA-binding protein, acts as an oncogene in medulloblastomas by regulating cellular survival and apoptosis. We observed different cellular responses upon YBX1 knockdown in several medulloblastoma cell lines, with significantly altered transcription and subsequent apoptosis rates. Mechanistically, PAR-CLIP for YBX1 and integration with RNA-Seq data uncovered direct posttranscriptional control of the heterochromatin-associated gene CBX5; upon YBX1 knockdown and subsequent CBX5 mRNA instability, heterochromatin-regulated genes involved in inflammatory response, apoptosis and death receptor signaling were de-repressed. Thus, YBX1 acts as an oncogene in medulloblastoma through indirect transcriptional regulation of inflammatory genes regulating apoptosis and represents a promising novel therapeutic target in this tumor entity.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Neoplastic , Heterochromatin/genetics , Inflammation/pathology , Medulloblastoma/pathology , RNA, Messenger/metabolism , Y-Box-Binding Protein 1/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Medulloblastoma/genetics , Medulloblastoma/immunology , Medulloblastoma/metabolism , RNA, Messenger/genetics , Tumor Cells, Cultured , Y-Box-Binding Protein 1/genetics
11.
Nat Neurosci ; 23(7): 842-853, 2020 07.
Article in English | MEDLINE | ID: mdl-32424282

ABSTRACT

Many immunotherapies act by enhancing the ability of cytotoxic T cells to kill tumor cells. Killing depends on T cell recognition of antigens presented by class I major histocompatibility complex (MHC-I) proteins on tumor cells. In this study, we showed that medulloblastomas lacking the p53 tumor suppressor do not express surface MHC-I and are therefore resistant to immune rejection. Mechanistically, this is because p53 regulates expression of the peptide transporter Tap1 and the aminopeptidase Erap1, which are required for MHC-I trafficking to the cell surface. In vitro, tumor necrosis factor (TNF) or lymphotoxin-ß receptor agonist can rescue expression of Erap1, Tap1 and MHC-I on p53-mutant tumor cells. In vivo, low doses of TNF prolong survival and synergize with immune checkpoint inhibitors to promote tumor rejection. These studies identified p53 as a key regulator of immune evasion and suggest that TNF could be used to enhance sensitivity of tumors to immunotherapy.


Subject(s)
Cerebellar Neoplasms/immunology , Medulloblastoma/immunology , Tumor Escape/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Protein p53/immunology , Animals , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Nat Med ; 26(5): 720-731, 2020 05.
Article in English | MEDLINE | ID: mdl-32341580

ABSTRACT

Recurrent medulloblastoma and ependymoma are universally lethal, with no approved targeted therapies and few candidates presently under clinical evaluation. Nearly all recurrent medulloblastomas and posterior fossa group A (PFA) ependymomas are located adjacent to and bathed by the cerebrospinal fluid, presenting an opportunity for locoregional therapy, bypassing the blood-brain barrier. We identify three cell-surface targets, EPHA2, HER2 and interleukin 13 receptor α2, expressed on medulloblastomas and ependymomas, but not expressed in the normal developing brain. We validate intrathecal delivery of EPHA2, HER2 and interleukin 13 receptor α2 chimeric antigen receptor T cells as an effective treatment for primary, metastatic and recurrent group 3 medulloblastoma and PFA ependymoma xenografts in mouse models. Finally, we demonstrate that administration of these chimeric antigen receptor T cells into the cerebrospinal fluid, alone or in combination with azacytidine, is a highly effective therapy for multiple metastatic mouse models of group 3 medulloblastoma and PFA ependymoma, thereby providing a rationale for clinical trials of these approaches in humans.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cerebrospinal Fluid/drug effects , Ependymoma/therapy , Immunotherapy, Adoptive/methods , Medulloblastoma/therapy , Animals , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cerebellar Neoplasms/cerebrospinal fluid , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/therapy , Cerebrospinal Fluid/immunology , Child , Child, Preschool , Drug Delivery Systems/methods , Ependymoma/cerebrospinal fluid , Ependymoma/immunology , Ependymoma/pathology , Female , HEK293 Cells , Humans , Infant , Injections, Intraventricular , Male , Medulloblastoma/cerebrospinal fluid , Medulloblastoma/immunology , Medulloblastoma/pathology , Mice , Neoplasm Metastasis , Receptors, Chimeric Antigen/administration & dosage , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
J Neuroimmunol ; 341: 577184, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32058173

ABSTRACT

We present an illustrative case of a 62-year-old woman with small cell lung cancer who developed progressive worsening of pre-existing anti-Hu antibody associated sensory neuronopathy after treatment with programmed cell death-1 (PD-1) inhibitor, nivolumab. We review the literature and identify 6 reported cases to understand the clinical outcomes of patients with anti-Hu paraneoplastic neurologic syndrome (PNS) treated with anti-PD-1 treatment. The PNS clinical spectrum comprised of encephalitis, a combination of sensory neuronopathy and anti-NMDAR encephalitis, isolated sensory neuronopathy, and encephalomyelitis. Immune checkpoint inhibitor have the potential to worsen pre-existing anti-Hu PNS and may promote the development of anti-Hu PNS.


Subject(s)
Antibodies, Antinuclear/blood , Antineoplastic Agents, Immunological/adverse effects , Autoantigens/immunology , ELAV Proteins/immunology , Nivolumab/adverse effects , Paraneoplastic Syndromes, Nervous System/etiology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Antinuclear/immunology , Antibodies, Neoplasm , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Carcinoma, Small Cell/complications , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/immunology , Carcinoma, Small Cell/secondary , Cerebellar Neoplasms/complications , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/secondary , Combined Modality Therapy , Disease Progression , Etoposide/administration & dosage , Female , Humans , Lung Neoplasms/complications , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/radiotherapy , Middle Aged , Nivolumab/therapeutic use , Paraneoplastic Syndromes, Nervous System/chemically induced , Paraneoplastic Syndromes, Nervous System/immunology , Radiotherapy, Adjuvant , Treatment Outcome
14.
Cancer Gene Ther ; 27(3-4): 246-255, 2020 04.
Article in English | MEDLINE | ID: mdl-30918335

ABSTRACT

Primary malignant central nervous system (CNS) tumors are the leading cause of childhood cancer-related death and morbidity. While advances in surgery, radiation, and chemotherapy have improved the survival rates in children with malignant brain tumors, mortality persists in certain subpopulations and current therapies are associated with extreme morbidity. This is especially true for children with malignant infratentorial tumors. Accordingly, G207, a genetically engineered herpes simplex virus (HSV-1) capable of selectively targeting cancer cells has emerged as a promising therapeutic option for this patient population. Herein, we demonstrate that cerebellar inoculation of G207 was systemically non-toxic in an immunocompetent, HSV-1 sensitive mouse strain (CBA/J). Mice had neither abnormal brain/organ pathology nor evidence of G207 replication by immunohistochemistry at days 7 and 30 after cerebellar G207 inoculation. While a minute amount viral DNA was recovered in the cerebellum and brainstem of mice at day 7, no viral DNA persisted at day 30. Critically, G207 delivered to the cerebellum was able to target/treat the highly aggressive MYC-overexpressed group 3 murine medulloblastoma increasing survival vs controls. These results provide critical safety and efficacy data to support the translation of G207 for pediatric clinical trials in intractable cerebellar malignancies.


Subject(s)
Cerebellar Neoplasms/therapy , Herpesvirus 1, Human/immunology , Medulloblastoma/therapy , Oncolytic Virotherapy/methods , Animals , Brain Stem/pathology , Brain Stem/virology , Cell Line, Tumor/transplantation , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Cerebellum/pathology , Cerebellum/virology , DNA, Viral/isolation & purification , Disease Models, Animal , Genetic Engineering , Herpesvirus 1, Human/genetics , Humans , Injections, Intralesional , Medulloblastoma/immunology , Medulloblastoma/pathology , Mice , Mice, Inbred CBA , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology
15.
Sci Adv ; 5(11): eaav9879, 2019 11.
Article in English | MEDLINE | ID: mdl-31807694

ABSTRACT

In both human and murine systems, we have developed an adoptive cellular therapy platform against medulloblastoma and glioblastoma that uses dendritic cells pulsed with a tumor RNA transcriptome to expand polyclonal tumor-reactive T cells against a plurality of antigens within heterogeneous brain tumors. We demonstrate that peripheral TCR Vß repertoire analysis after adoptive cellular therapy reveals that effective response to adoptive cellular therapy is concordant with massive in vivo expansion and persistence of tumor-specific T cell clones within the peripheral blood. In preclinical models of medulloblastoma and glioblastoma, and in a patient with relapsed medulloblastoma receiving adoptive cellular therapy, an early and massive expansion of tumor-reactive lymphocytes, coupled with prolonged persistence in the peripheral blood, is observed during effective therapeutic response to immunotherapy treatment.


Subject(s)
Adoptive Transfer , Cerebellar Neoplasms , Immunotherapy, Adoptive , Medulloblastoma , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes , Animals , Cell Line, Tumor , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/therapy , Humans , Medulloblastoma/immunology , Medulloblastoma/pathology , Medulloblastoma/therapy , Mice , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/pathology
16.
J Transl Med ; 17(1): 321, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31547819

ABSTRACT

BACKGROUND: Medulloblastoma (MB), the most common pediatric brain cancer, presents with a poor prognosis in a subset of patients with high risk disease, or at recurrence, where current therapies are ineffective. Cord blood (CB) natural killer (NK) cells may be promising off-the-shelf effector cells for immunotherapy due to their recognition of malignant cells without the need for a known target, ready availability from multiple banks, and their potential to expand exponentially. However, they are currently limited by immune suppressive cytokines secreted in the MB tumor microenvironment including Transforming Growth Factor ß (TGF-ß). Here, we address this challenge in in vitro models of MB. METHODS: CB-derived NK cells were modified to express a dominant negative TGF-ß receptor II (DNRII) using retroviral transduction. The ability of transduced CB cells to maintain function in the presence of medulloblastoma-conditioned media was then assessed. RESULTS: We observed that the cytotoxic ability of nontransduced CB-NK cells was reduced in the presence of TGF-ß-rich, medulloblastoma-conditioned media (21.21 ± 1.19% killing at E:T 5:1 in the absence vs. 14.98 ± 2.11% in the presence of medulloblastoma-conditioned media, n = 8, p = 0.02), but was unaffected in CB-derived DNRII-transduced NK cells (21.11 ± 1.84% killing at E:T 5:1 in the absence vs. 21.81 ± 3.37 in the presence of medulloblastoma-conditioned media, n = 8, p = 0.85. We also observed decreased expression of CCR2 in untransduced NK cells (mean CCR2 MFI 826 ± 117 in untransduced NK + MB supernatant from mean CCR2 MFI 1639.29 ± 215 in no MB supernatant, n = 7, p = 0.0156), but not in the transduced cells. Finally, we observed that CB-derived DNRII-transduced NK cells may protect surrounding immune cells by providing a cytokine sink for TGF-ß (decreased TGF-ß levels of 610 ± 265 pg/mL in CB-derived DNRII-transduced NK cells vs. 1817 ± 342 pg/mL in untransduced cells; p = 0.008). CONCLUSIONS: CB NK cells expressing a TGF-ß DNRII may have a functional advantage over unmodified NK cells in the presence of TGF-ß-rich MB, warranting further investigation on its potential applications for patients with medulloblastoma.


Subject(s)
Cerebellar Neoplasms/immunology , Killer Cells, Natural/immunology , Medulloblastoma/immunology , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Down-Regulation , Fetal Blood/cytology , Humans , Killer Cells, Natural/transplantation , Neutralization Tests , Receptors, CCR2/metabolism , Transplantation, Homologous
17.
Nat Commun ; 10(1): 2410, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160587

ABSTRACT

Medulloblastoma, which is the most common malignant paediatric brain tumour, has a 70% survival rate, but standard treatments often lead to devastating life-long side effects and recurrence is fatal. One of the emerging strategies in the search for treatments is to determine the roles of tumour microenvironment cells in the growth and maintenance of tumours. The most attractive target is tumour-associated macrophages (TAMs), which are abundantly present in the Sonic Hedgehog (SHH) subgroup of medulloblastoma. Here, we report an unexpected beneficial role of TAMs in SHH medulloblastoma. In human patients, decreased macrophage number is correlated with significantly poorer outcome. We confirm macrophage anti-tumoural behaviour in both ex vivo and in vivo murine models of SHH medulloblastoma. Taken together, our findings suggest that macrophages play a positive role by impairing tumour growth in medulloblastoma, in contrast to the pro-tumoural role played by TAMs in glioblastoma, another common brain tumour.


Subject(s)
Cerebellar Neoplasms/immunology , Macrophages/immunology , Medulloblastoma/immunology , Tumor Microenvironment/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , Calcium-Binding Proteins , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Hedgehog Proteins/metabolism , Humans , Macrophages/metabolism , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Microfilament Proteins , Microglia/immunology , Myeloid Cells/immunology , Receptors, CCR2/genetics , Up-Regulation
18.
J Neurooncol ; 142(3): 395-407, 2019 May.
Article in English | MEDLINE | ID: mdl-30788681

ABSTRACT

PURPOSE: Medulloblastoma (MB) is the most common malignant brain tumor in children. Recent studies have shown the ability of natural killer (NK) cells to lyse MB cell lines in vitro, but in vivo successes remain elusive and the efficacy and fate of NK cells in vivo remain unknown. METHODS: To address these questions, we injected MB cells into the cerebellum of immunodeficient mice and examined tumor growth at various days after tumor establishment via bioluminescence imaging. NK cells were labeled with a fluorine-19 (19F) MRI probe and subsequently injected either intratumorally or contralaterally to the tumor in the cerebellum and effect on tumor growth was monitored. RESULTS: The 19F probe efficiently labeled the NK cells and exhibited little cytotoxicity. Fluorine-19 MRI confirmed the successful and accurate delivery of the labeled NK cells to the cerebellum of the mice. Administration of 19F-labeled NK cells suppressed MB growth, with the same efficacy as unlabeled cells. Immunohistochemistry confirmed the presence of NK cells within the tumor, which was associated with induction of apoptosis in tumor cells. NK cell migration to the tumor from a distal location as well as activation of apoptosis was also demonstrated by immunohstochemistry. CONCLUSIONS: Our results show that NK cells present a novel opportunity for new strategies in MB treatment. Further, 19F-labeled NK cells can suppress MB growth while enabling 19F MRI to provide imaging feedback that can facilitate study and optimization of therapeutic paradigms.


Subject(s)
Cerebellar Neoplasms/prevention & control , Drug Monitoring/methods , Fluorine Radioisotopes/therapeutic use , Killer Cells, Natural/transplantation , Magnetic Resonance Imaging/methods , Medulloblastoma/prevention & control , Animals , Apoptosis , Cell Proliferation , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Humans , Medulloblastoma/immunology , Medulloblastoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Neuro Oncol ; 20(12): 1606-1615, 2018 11 12.
Article in English | MEDLINE | ID: mdl-29912438

ABSTRACT

Background: Oncolytic measles virus (MV) is effective in xenograft models of many tumor types in immune-compromised mice. However, no murine cell line exists that is tumorigenic, grows in immune-competent mice, and is killed by MV. The lack of such a model prevents an examination of the effect of the immune system on MV oncotherapy. Methods: Cerebellar stem cells from human CD46-transgenic immunocompetent mice were transduced to express Sendai virus C-protein, murine C-Myc, and Gfi1b proteins. The resultant cells were injected into the brain of NSG mice, and a cell line, called CSCG, was prepared from the resulting tumor. Results: CSCG cells are highly proliferative, and express stem cell markers. These cells are permissive for replication of MV and are killed by the virus in a dose- and time-dependent manner. CSCG cells form aggressive tumors that morphologically resemble medulloblastoma when injected into the brains of immune-competent mice. On the molecular level, CSCG tumors overexpress natriuretic peptide receptor 3 and gamma-aminobutyric acid type A receptor alpha 5, markers of Group 3 medulloblastoma. A single intratumoral injection of MV‒green fluorescent protein resulted in complete tumor regression and prolonged survival of animals compared with treatments with phosphate buffered saline (P = 0.0018) or heat-inactivated MV (P = 0.0027). Conclusions: This immune-competent model provides the first platform to test therapeutic regimens of oncolytic MV for Group 3 medulloblastoma in the presence of anti-measles immunity. The strategy presented here can be used to make MV-sensitive murine models of any human tumor for which the driving mutations are known.


Subject(s)
Cerebellar Neoplasms/therapy , Disease Models, Animal , Immunocompetence , Measles virus/genetics , Medulloblastoma/therapy , Oncolytic Virotherapy , Animals , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/virology , Humans , Measles/virology , Medulloblastoma/immunology , Medulloblastoma/metabolism , Medulloblastoma/virology , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Cells, Cultured , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Xenograft Model Antitumor Assays
20.
BMC Cancer ; 18(1): 535, 2018 May 08.
Article in English | MEDLINE | ID: mdl-29739450

ABSTRACT

BACKGROUND: Recent progress in molecular analysis has advanced the understanding of medulloblastoma (MB) and is anticipated to facilitate management of the disease. MB is composed of 4 molecular subgroups: WNT, SHH, Group 3, and Group 4. Macrophages play a crucial role in the tumor microenvironment; however, the functional role of their activated phenotype (M1/M2) remains controversial. Herein, we investigate the correlation between tumor-associated macrophage (TAM) recruitment within the MB subgroups and prognosis. METHODS: Molecular subgrouping was performed by a nanoString-based RNA assay on retrieved snap-frozen tissue samples. Immunohistochemistry (IHC) and immunofluorescence (IF) assays were performed on subgroup identified samples, and the number of polarized macrophages was quantified from IHC. Survival analyses were conducted on collected clinical data and quantified macrophage data. RESULTS: TAM (M1/M2) recruitment in SHH MB was significantly higher compared to that in other subgroups. A Kaplan-Meier survival curve and multivariate Cox regression demonstrated that high M1 expressers showed worse overall survival (OS) and progression-free survival (PFS) than low M1 expressers in SHH MB, with relative risk (RR) values of 11.918 and 6.022, respectively. CONCLUSION: M1 rather than M2 correlates more strongly with worse outcome in SHH medulloblastoma.


Subject(s)
Cerebellar Neoplasms/immunology , Hedgehog Proteins/metabolism , Macrophages/immunology , Medulloblastoma/immunology , Tumor Microenvironment/immunology , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Female , Gene Expression Profiling , Humans , Male , Medulloblastoma/mortality , Medulloblastoma/pathology , Prognosis , Progression-Free Survival , Survival Analysis , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...