Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38971152

RESUMEN

We identify a population of Protogenin-positive (PRTG+ve) MYChigh NESTINlow stem cells in the four-week-old human embryonic hindbrain that subsequently localizes to the ventricular zone of the rhombic lip (RLVZ). Oncogenic transformation of early Prtg+ve rhombic lip stem cells initiates group 3 medulloblastoma (Gr3-MB)-like tumors. PRTG+ve stem cells grow adjacent to a human-specific interposed vascular plexus in the RLVZ, a phenotype that is recapitulated in Gr3-MB but not in other types of medulloblastoma. Co-culture of Gr3-MB with endothelial cells promotes tumor stem cell growth, with the endothelial cells adopting an immature phenotype. Targeting the PRTGhigh compartment of Gr3-MB in vivo using either the diphtheria toxin system or chimeric antigen receptor T cells constitutes effective therapy. Human Gr3-MBs likely arise from early embryonic RLVZ PRTG+ve stem cells inhabiting a specific perivascular niche. Targeting the PRTGhigh compartment and/or the perivascular niche represents an approach to treat children with Gr3-MB.

3.
Nat Commun ; 12(1): 1749, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741928

RESUMEN

Sonic hedgehog medulloblastoma encompasses a clinically and molecularly diverse group of cancers of the developing central nervous system. Here, we use unbiased sequencing of the transcriptome across a large cohort of 250 tumors to reveal differences among molecular subtypes of the disease, and demonstrate the previously unappreciated importance of non-coding RNA transcripts. We identify alterations within the cAMP dependent pathway (GNAS, PRKAR1A) which converge on GLI2 activity and show that 18% of tumors have a genetic event that directly targets the abundance and/or stability of MYCN. Furthermore, we discover an extensive network of fusions in focally amplified regions encompassing GLI2, and several loss-of-function fusions in tumor suppressor genes PTCH1, SUFU and NCOR1. Molecular convergence on a subset of genes by nucleotide variants, copy number aberrations, and gene fusions highlight the key roles of specific pathways in the pathogenesis of Sonic hedgehog medulloblastoma and open up opportunities for therapeutic intervention.


Asunto(s)
Neoplasias Cerebelosas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Meduloblastoma/genética , Transcriptoma , Adolescente , Adulto , Niño , Preescolar , Femenino , Redes Reguladoras de Genes , Variación Genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Transducción de Señal/genética , Adulto Joven
4.
Cell ; 181(6): 1329-1345.e24, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32445698

RESUMEN

Posterior fossa A (PFA) ependymomas are lethal malignancies of the hindbrain in infants and toddlers. Lacking highly recurrent somatic mutations, PFA ependymomas are proposed to be epigenetically driven tumors for which model systems are lacking. Here we demonstrate that PFA ependymomas are maintained under hypoxia, associated with restricted availability of specific metabolites to diminish histone methylation, and increase histone demethylation and acetylation at histone 3 lysine 27 (H3K27). PFA ependymomas initiate from a cell lineage in the first trimester of human development that resides in restricted oxygen. Unlike other ependymomas, transient exposure of PFA cells to ambient oxygen induces irreversible cellular toxicity. PFA tumors exhibit a low basal level of H3K27me3, and, paradoxically, inhibition of H3K27 methylation specifically disrupts PFA tumor growth. Targeting metabolism and/or the epigenome presents a unique opportunity for rational therapy for infants with PFA ependymoma.


Asunto(s)
Ependimoma/genética , Ependimoma/metabolismo , Epigenoma/genética , Neoplasias Infratentoriales/genética , Neoplasias Infratentoriales/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular , Proliferación Celular/genética , Metilación de ADN/genética , Epigenómica/métodos , Histonas/genética , Histonas/metabolismo , Humanos , Lactante , Lisina/genética , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Mutación/genética
5.
Nat Med ; 26(5): 720-731, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32341580

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/administración & dosificación , Líquido Cefalorraquídeo/efectos de los fármacos , Ependimoma/terapia , Inmunoterapia Adoptiva/métodos , Meduloblastoma/terapia , Animales , Neoplasias Encefálicas/líquido cefalorraquídeo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Cerebelosas/líquido cefalorraquídeo , Neoplasias Cerebelosas/inmunología , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/terapia , Líquido Cefalorraquídeo/inmunología , Niño , Preescolar , Sistemas de Liberación de Medicamentos/métodos , Ependimoma/líquido cefalorraquídeo , Ependimoma/inmunología , Ependimoma/patología , Femenino , Células HEK293 , Humanos , Lactante , Inyecciones Intraventriculares , Masculino , Meduloblastoma/líquido cefalorraquídeo , Meduloblastoma/inmunología , Meduloblastoma/patología , Ratones , Metástasis de la Neoplasia , Receptores Quiméricos de Antígenos/administración & dosificación , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Resultado del Tratamiento , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Nature ; 574(7780): 707-711, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31664194

RESUMEN

In cancer, recurrent somatic single-nucleotide variants-which are rare in most paediatric cancers-are confined largely to protein-coding genes1-3. Here we report highly recurrent hotspot mutations (r.3A>G) of U1 spliceosomal small nuclear RNAs (snRNAs) in about 50% of Sonic hedgehog (SHH) medulloblastomas. These mutations were not present across other subgroups of medulloblastoma, and we identified these hotspot mutations in U1 snRNA in only <0.1% of 2,442 cancers, across 36 other tumour types. The mutations occur in 97% of adults (subtype SHHδ) and 25% of adolescents (subtype SHHα) with SHH medulloblastoma, but are largely absent from SHH medulloblastoma in infants. The U1 snRNA mutations occur in the 5' splice-site binding region, and snRNA-mutant tumours have significantly disrupted RNA splicing and an excess of 5' cryptic splicing events. Alternative splicing mediated by mutant U1 snRNA inactivates tumour-suppressor genes (PTCH1) and activates oncogenes (GLI2 and CCND2), and represents a target for therapy. These U1 snRNA mutations provide an example of highly recurrent and tissue-specific mutations of a non-protein-coding gene in cancer.


Asunto(s)
Neoplasias Cerebelosas/genética , Proteínas Hedgehog/genética , Meduloblastoma/genética , ARN Nuclear Pequeño/genética , Adolescente , Adulto , Empalme Alternativo , Proteínas Hedgehog/metabolismo , Humanos , Mutación , Sitios de Empalme de ARN , Empalme del ARN
7.
Nature ; 572(7767): 67-73, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31043743

RESUMEN

Study of the origin and development of cerebellar tumours has been hampered by the complexity and heterogeneity of cerebellar cells that change over the course of development. Here we use single-cell transcriptomics to study more than 60,000 cells from the developing mouse cerebellum and show that different molecular subgroups of childhood cerebellar tumours mirror the transcription of cells from distinct, temporally restricted cerebellar lineages. The Sonic Hedgehog medulloblastoma subgroup transcriptionally mirrors the granule cell hierarchy as expected, while group 3 medulloblastoma resembles Nestin+ stem cells, group 4 medulloblastoma resembles unipolar brush cells, and PFA/PFB ependymoma and cerebellar pilocytic astrocytoma resemble the prenatal gliogenic progenitor cells. Furthermore, single-cell transcriptomics of human childhood cerebellar tumours demonstrates that many bulk tumours contain a mixed population of cells with divergent differentiation. Our data highlight cerebellar tumours as a disorder of early brain development and provide a proximate explanation for the peak incidence of cerebellar tumours in early childhood.


Asunto(s)
Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Evolución Molecular , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Transcripción Genética , Animales , Neoplasias Cerebelosas/clasificación , Cerebelo/citología , Cerebelo/embriología , Cerebelo/metabolismo , Niño , Femenino , Feto/citología , Glioma/clasificación , Glioma/genética , Glioma/patología , Humanos , Meduloblastoma/clasificación , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Factores de Tiempo , Transcriptoma/genética
8.
Cancer Res ; 79(5): 905-917, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30674530

RESUMEN

Medulloblastoma and central nervous system primitive neuroectodermal tumors (CNS-PNET) are aggressive, poorly differentiated brain tumors with limited effective therapies. Using Sleeping Beauty (SB) transposon mutagenesis, we identified novel genetic drivers of medulloblastoma and CNS-PNET. Cross-species gene expression analyses classified SB-driven tumors into distinct medulloblastoma and CNS-PNET subgroups, indicating they resemble human Sonic hedgehog and group 3 and 4 medulloblastoma and CNS neuroblastoma with FOXR2 activation. This represents the first genetically induced mouse model of CNS-PNET and a rare model of group 3 and 4 medulloblastoma. We identified several putative proto-oncogenes including Arhgap36, Megf10, and Foxr2. Genetic manipulation of these genes demonstrated a robust impact on tumorigenesis in vitro and in vivo. We also determined that FOXR2 interacts with N-MYC, increases C-MYC protein stability, and activates FAK/SRC signaling. Altogether, our study identified several promising therapeutic targets in medulloblastoma and CNS-PNET. SIGNIFICANCE: A transposon-induced mouse model identifies several novel genetic drivers and potential therapeutic targets in medulloblastoma and CNS-PNET.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Cerebelosas/genética , Meduloblastoma/genética , Tumores Neuroectodérmicos Primitivos/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/genética , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Elementos Transponibles de ADN/genética , Femenino , Factores de Transcripción Forkhead/genética , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , Humanos , Masculino , Meduloblastoma/metabolismo , Meduloblastoma/patología , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Mutagénesis Insercional/métodos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Tumores Neuroectodérmicos Primitivos/metabolismo , Tumores Neuroectodérmicos Primitivos/patología , Pronóstico
11.
Cell ; 172(5): 1050-1062.e14, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474906

RESUMEN

While the preponderance of morbidity and mortality in medulloblastoma patients are due to metastatic disease, most research focuses on the primary tumor due to a dearth of metastatic tissue samples and model systems. Medulloblastoma metastases are found almost exclusively on the leptomeningeal surface of the brain and spinal cord; dissemination is therefore thought to occur through shedding of primary tumor cells into the cerebrospinal fluid followed by distal re-implantation on the leptomeninges. We present evidence for medulloblastoma circulating tumor cells (CTCs) in therapy-naive patients and demonstrate in vivo, through flank xenografting and parabiosis, that medulloblastoma CTCs can spread through the blood to the leptomeningeal space to form leptomeningeal metastases. Medulloblastoma leptomeningeal metastases express high levels of the chemokine CCL2, and expression of CCL2 in medulloblastoma in vivo is sufficient to drive leptomeningeal dissemination. Hematogenous dissemination of medulloblastoma offers a new opportunity to diagnose and treat lethal disseminated medulloblastoma.


Asunto(s)
Meduloblastoma/irrigación sanguínea , Meduloblastoma/patología , Neoplasias Meníngeas/irrigación sanguínea , Neoplasias Meníngeas/secundario , Aloinjertos , Animales , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Cromosomas Humanos Par 10/genética , Femenino , Humanos , Masculino , Meduloblastoma/genética , Ratones SCID , Células Neoplásicas Circulantes , Parabiosis
12.
Neuro Oncol ; 20(3): 313-323, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-28575493

RESUMEN

microRNAs (miRNAs) have wide-ranging effects on large-scale gene regulation. As such, they play a vital role in dictating normal development, and their aberrant expression has been implicated in cancer. There has been a large body of research on the role of miRNAs in medulloblastoma, the most common malignant brain tumor of childhood. The identification of the 4 molecular subgroups with distinct biological, genetic, and transcriptional features has revolutionized the field of medulloblastoma research over the past 5 years. Despite this, the growing body of research on miRNAs in medulloblastoma has largely focused on the clinical entity of a single disease rather than the molecular subgroups. This review begins by highlighting the role of miRNAs in development and progresses to explore their myriad of implications in cancer. Medulloblastoma is characterized by increased proliferation, inhibition of apoptosis, and maintenance of stemness programs-features that are inadvertently regulated by altered expression patterns in miRNAs. This review aims to contextualize the large body of work on miRNAs within the framework of medulloblastoma subgroups. The goal of this review is to stimulate new areas of research, including potential therapeutics, within a rapidly growing field.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Cerebelosas/diagnóstico , Regulación Neoplásica de la Expresión Génica , Meduloblastoma/diagnóstico , MicroARNs/genética , Neoplasias Cerebelosas/genética , Humanos , Meduloblastoma/genética , Pronóstico
13.
Nature ; 553(7686): 101-105, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29258295

RESUMEN

Genomic sequencing has driven precision-based oncology therapy; however, the genetic drivers of many malignancies remain unknown or non-targetable, so alternative approaches to the identification of therapeutic leads are necessary. Ependymomas are chemotherapy-resistant brain tumours, which, despite genomic sequencing, lack effective molecular targets. Intracranial ependymomas are segregated on the basis of anatomical location (supratentorial region or posterior fossa) and further divided into distinct molecular subgroups that reflect differences in the age of onset, gender predominance and response to therapy. The most common and aggressive subgroup, posterior fossa ependymoma group A (PF-EPN-A), occurs in young children and appears to lack recurrent somatic mutations. Conversely, posterior fossa ependymoma group B (PF-EPN-B) tumours display frequent large-scale copy number gains and losses but have favourable clinical outcomes. More than 70% of supratentorial ependymomas are defined by highly recurrent gene fusions in the NF-κB subunit gene RELA (ST-EPN-RELA), and a smaller number involve fusion of the gene encoding the transcriptional activator YAP1 (ST-EPN-YAP1). Subependymomas, a distinct histologic variant, can also be found within the supratetorial and posterior fossa compartments, and account for the majority of tumours in the molecular subgroups ST-EPN-SE and PF-EPN-SE. Here we describe mapping of active chromatin landscapes in 42 primary ependymomas in two non-overlapping primary ependymoma cohorts, with the goal of identifying essential super-enhancer-associated genes on which tumour cells depend. Enhancer regions revealed putative oncogenes, molecular targets and pathways; inhibition of these targets with small molecule inhibitors or short hairpin RNA diminished the proliferation of patient-derived neurospheres and increased survival in mouse models of ependymomas. Through profiling of transcriptional enhancers, our study provides a framework for target and drug discovery in other cancers that lack known genetic drivers and are therefore difficult to treat.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Ependimoma/tratamiento farmacológico , Ependimoma/genética , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes/genética , Terapia Molecular Dirigida , Oncogenes/genética , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Ependimoma/clasificación , Ependimoma/patología , Femenino , Humanos , Ratones , Medicina de Precisión , Interferencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nature ; 547(7663): 311-317, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28726821

RESUMEN

Current therapies for medulloblastoma, a highly malignant childhood brain tumour, impose debilitating effects on the developing child, and highlight the need for molecularly targeted treatments with reduced toxicity. Previous studies have been unable to identify the full spectrum of driver genes and molecular processes that operate in medulloblastoma subgroups. Here we analyse the somatic landscape across 491 sequenced medulloblastoma samples and the molecular heterogeneity among 1,256 epigenetically analysed cases, and identify subgroup-specific driver alterations that include previously undiscovered actionable targets. Driver mutations were confidently assigned to most patients belonging to Group 3 and Group 4 medulloblastoma subgroups, greatly enhancing previous knowledge. New molecular subtypes were differentially enriched for specific driver events, including hotspot in-frame insertions that target KBTBD4 and 'enhancer hijacking' events that activate PRDM6. Thus, the application of integrative genomics to an extensive cohort of clinical samples derived from a single childhood cancer entity revealed a series of cancer genes and biologically relevant subtype diversity that represent attractive therapeutic targets for the treatment of patients with medulloblastoma.


Asunto(s)
Análisis Mutacional de ADN , Genoma Humano/genética , Meduloblastoma/clasificación , Meduloblastoma/genética , Secuenciación Completa del Genoma , Carcinogénesis/genética , Proteínas Portadoras/genética , Estudios de Cohortes , Metilación de ADN , Conjuntos de Datos como Asunto , Epistasis Genética , Genómica , Humanos , Terapia Molecular Dirigida , Proteínas Musculares/genética , Mutación , Oncogenes/genética , Factores de Transcripción/genética , Proteínas Wnt/genética
15.
Nat Genet ; 49(5): 780-788, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28394352

RESUMEN

Spatial heterogeneity of transcriptional and genetic markers between physically isolated biopsies of a single tumor poses major barriers to the identification of biomarkers and the development of targeted therapies that will be effective against the entire tumor. We analyzed the spatial heterogeneity of multiregional biopsies from 35 patients, using a combination of transcriptomic and genomic profiles. Medulloblastomas (MBs), but not high-grade gliomas (HGGs), demonstrated spatially homogeneous transcriptomes, which allowed for accurate subgrouping of tumors from a single biopsy. Conversely, somatic mutations that affect genes suitable for targeted therapeutics demonstrated high levels of spatial heterogeneity in MB, malignant glioma, and renal cell carcinoma (RCC). Actionable targets found in a single MB biopsy were seldom clonal across the entire tumor, which brings the efficacy of monotherapies against a single target into question. Clinical trials of targeted therapies for MB should first ensure the spatially ubiquitous nature of the target mutation.


Asunto(s)
Neoplasias Cerebelosas/genética , Regulación Neoplásica de la Expresión Génica , Meduloblastoma/genética , Transcriptoma , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Cerebelosas/patología , Niño , Preescolar , Análisis por Conglomerados , Variaciones en el Número de Copia de ADN , Femenino , Perfilación de la Expresión Génica/métodos , Heterogeneidad Genética , Estudio de Asociación del Genoma Completo , Humanos , Mutación INDEL , Masculino , Meduloblastoma/patología , Persona de Mediana Edad , Mutación , Polimorfismo de Nucleótido Simple , Análisis de Componente Principal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Nature ; 529(7586): 351-7, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26760213

RESUMEN

The development of targeted anti-cancer therapies through the study of cancer genomes is intended to increase survival rates and decrease treatment-related toxicity. We treated a transposon-driven, functional genomic mouse model of medulloblastoma with 'humanized' in vivo therapy (microneurosurgical tumour resection followed by multi-fractionated, image-guided radiotherapy). Genetic events in recurrent murine medulloblastoma exhibit a very poor overlap with those in matched murine diagnostic samples (<5%). Whole-genome sequencing of 33 pairs of human diagnostic and post-therapy medulloblastomas demonstrated substantial genetic divergence of the dominant clone after therapy (<12% diagnostic events were retained at recurrence). In both mice and humans, the dominant clone at recurrence arose through clonal selection of a pre-existing minor clone present at diagnosis. Targeted therapy is unlikely to be effective in the absence of the target, therefore our results offer a simple, proximal, and remediable explanation for the failure of prior clinical trials of targeted therapy.


Asunto(s)
Neoplasias Cerebelosas/terapia , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Meduloblastoma/terapia , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Selección Genética/efectos de los fármacos , Animales , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/radioterapia , Neoplasias Cerebelosas/cirugía , Células Clonales/patología , Irradiación Craneoespinal , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Femenino , Genoma Humano/genética , Humanos , Masculino , Meduloblastoma/genética , Meduloblastoma/patología , Meduloblastoma/radioterapia , Meduloblastoma/cirugía , Ratones , Terapia Molecular Dirigida/métodos , Recurrencia Local de Neoplasia/terapia , Radioterapia Guiada por Imagen , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Nat Neurosci ; 18(9): 1236-46, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26258683

RESUMEN

Over 20% of the drugs for treating human diseases target ion channels, but no cancer drug approved by the US Food and Drug Administration (FDA) is intended to target an ion channel. We found that the EAG2 (Ether-a-go-go 2) potassium channel has an evolutionarily conserved function for promoting brain tumor growth and metastasis, delineate downstream pathways, and uncover a mechanism for different potassium channels to functionally cooperate and regulate mitotic cell volume and tumor progression. EAG2 potassium channel was enriched at the trailing edge of migrating medulloblastoma (MB) cells to regulate local cell volume dynamics, thereby facilitating cell motility. We identified the FDA-approved antipsychotic drug thioridazine as an EAG2 channel blocker that reduces xenografted MB growth and metastasis, and present a case report of repurposing thioridazine for treating a human patient. Our findings illustrate the potential of targeting ion channels in cancer treatment.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/fisiología , Evolución Molecular , Tioridazina/administración & dosificación , Animales , Neoplasias Encefálicas/diagnóstico , Células COS , Chlorocebus aethiops , Drosophila , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Adulto Joven
18.
Acta Neuropathol ; 129(3): 449-57, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25689980

RESUMEN

Medulloblastoma comprises four distinct molecular variants with distinct genetics, transcriptomes, and outcomes. Subgroup affiliation has been previously shown to remain stable at the time of recurrence, which likely reflects their distinct cells of origin. However, a therapeutically relevant question that remains unanswered is subgroup stability in the metastatic compartment. We assembled a cohort of 12-paired primary-metastatic tumors collected in the MAGIC consortium, and established their molecular subgroup affiliation by performing integrative gene expression and DNA methylation analysis. Frozen tissues were collected and profiled using Affymetrix gene expression arrays and Illumina methylation arrays. Class prediction and hierarchical clustering were performed using existing published datasets. Our molecular analysis, using consensus integrative genomic data, establishes the unequivocal maintenance of molecular subgroup affiliation in metastatic medulloblastoma. We further validated these findings by interrogating a non-overlapping cohort of 19 pairs of primary-metastatic tumors from the Burdenko Neurosurgical Institute using an orthogonal technique of immunohistochemical staining. This investigation represents the largest reported primary-metastatic paired cohort profiled to date and provides a unique opportunity to evaluate subgroup-specific molecular aberrations within the metastatic compartment. Our findings further support the hypothesis that medulloblastoma subgroups arise from distinct cells of origin, which are carried forward from ontogeny to oncology.


Asunto(s)
Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Meduloblastoma/genética , Meduloblastoma/patología , Metástasis de la Neoplasia/genética , Adolescente , Niño , Preescolar , Análisis por Conglomerados , Femenino , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcriptoma
19.
Cancer Res ; 75(1): 134-46, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25391241

RESUMEN

Medulloblastoma is the most common malignant pediatric brain tumor, with metastases present at diagnosis conferring a poor prognosis. Mechanisms of dissemination are poorly understood and metastatic lesions are genetically divergent from the matched primary tumor. Effective and less toxic therapies that target both compartments have yet to be identified. Here, we report that the analysis of several large nonoverlapping cohorts of patients with medulloblastoma reveals MET kinase as a marker of sonic hedgehog (SHH)-driven medulloblastoma. Immunohistochemical analysis of phosphorylated, active MET kinase in an independent patient cohort confirmed its correlation with increased tumor relapse and poor survival, suggesting that patients with SHH medulloblastoma may benefit from MET-targeted therapy. In support of this hypothesis, we found that the approved MET inhibitor foretinib could suppress MET activation, decrease tumor cell proliferation, and induce apoptosis in SHH medulloblastomas in vitro and in vivo. Foretinib penetrated the blood-brain barrier and was effective in both the primary and metastatic tumor compartments. In established mouse xenograft or transgenic models of metastatic SHH medulloblastoma, foretinib administration reduced the growth of the primary tumor, decreased the incidence of metastases, and increased host survival. Taken together, our results provide a strong rationale to clinically evaluate foretinib as an effective therapy for patients with SHH-driven medulloblastoma.


Asunto(s)
Anilidas/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/metabolismo , Proteínas Hedgehog/metabolismo , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/metabolismo , Quinolinas/farmacología , Anilidas/farmacocinética , Animales , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Perfilación de la Expresión Génica , Humanos , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Quinolinas/farmacocinética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Cell ; 26(6): 940-940.e1, 2014 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-25490452

RESUMEN

Medulloblastoma (MB) is the most common malignant brain tumor in children, where one-third of patients succumb to their disease. This SnapShot describes the classification of MB subgroups, historically by histopathology and currently based on genomic information. Genomics-based classification has identified four major subgroups and provides greater opportunity for developing targeted therapies more successful than current conventional therapy.


Asunto(s)
Neoplasias Cerebelosas/patología , Regulación Neoplásica de la Expresión Génica , Meduloblastoma/patología , Animales , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/terapia , Epigénesis Genética , Humanos , Meduloblastoma/genética , Meduloblastoma/terapia , Ratones , Neoplasias Experimentales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA