Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
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
2.
Nature ; 547(7663): 355-359, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28678782

RESUMEN

Glioblastoma is a universally lethal cancer with a median survival time of approximately 15 months. Despite substantial efforts to define druggable targets, there are no therapeutic options that notably extend the lifespan of patients with glioblastoma. While previous work has largely focused on in vitro cellular models, here we demonstrate a more physiologically relevant approach to target discovery in glioblastoma. We adapted pooled RNA interference (RNAi) screening technology for use in orthotopic patient-derived xenograft models, creating a high-throughput negative-selection screening platform in a functional in vivo tumour microenvironment. Using this approach, we performed parallel in vivo and in vitro screens and discovered that the chromatin and transcriptional regulators needed for cell survival in vivo are non-overlapping with those required in vitro. We identified transcription pause-release and elongation factors as one set of in vivo-specific cancer dependencies, and determined that these factors are necessary for enhancer-mediated transcriptional adaptations that enable cells to survive the tumour microenvironment. Our lead hit, JMJD6, mediates the upregulation of in vivo stress and stimulus response pathways through enhancer-mediated transcriptional pause-release, promoting cell survival specifically in vivo. Targeting JMJD6 or other identified elongation factors extends survival in orthotopic xenograft mouse models, suggesting that targeting transcription elongation machinery may be an effective therapeutic strategy for glioblastoma. More broadly, this study demonstrates the power of in vivo phenotypic screening to identify new classes of 'cancer dependencies' not identified by previous in vitro approaches, and could supply new opportunities for therapeutic intervention.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Terapia Molecular Dirigida/tendencias , Factores de Elongación Transcripcional/antagonistas & inhibidores , Factores de Elongación Transcripcional/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/metabolismo , Masculino , Ratones , Interferencia de ARN , Transcripción Genética , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Clin Invest ; 129(10): 4377-4392, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31498151

RESUMEN

Despite progress in intensification of therapy, outcomes for patients with metastatic osteosarcoma (OS) have not improved in thirty years. We developed a system that enabled preclinical screening of compounds against metastatic OS cells in the context of the native lung microenvironment. Using this strategy to screen a library of epigenetically targeted compounds, we identified inhibitors of CDK12 to be most effective, reducing OS cell outgrowth in the lung by more than 90% at submicromolar doses. We found that knockout of CDK12 in an in vivo model of lung metastasis significantly decreased the ability of OS to colonize the lung. CDK12 inhibition led to defects in transcription elongation in a gene length- and expression-dependent manner. These effects were accompanied by defects in RNA processing and altered the expression of genes involved in transcription regulation and the DNA damage response. We further identified OS models that differ in their sensitivity to CDK12 inhibition in the lung and provided evidence that upregulated MYC levels may mediate these differences. Our studies provided a framework for rapid preclinical testing of compounds with antimetastatic activity and highlighted CDK12 as a potential therapeutic target in OS.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Osteosarcoma/enzimología , Osteosarcoma/secundario , Animales , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/genética , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Técnicas de Inactivación de Genes , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Osteosarcoma/genética , Inhibidores de Proteínas Quinasas/farmacología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/fisiología
6.
Nat Med ; 24(2): 176-185, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29334376

RESUMEN

Metastasis results from a complex set of traits acquired by tumor cells, distinct from those necessary for tumorigenesis. Here, we investigate the contribution of enhancer elements to the metastatic phenotype of osteosarcoma. Through epigenomic profiling, we identify substantial differences in enhancer activity between primary and metastatic human tumors and between near isogenic pairs of highly lung metastatic and nonmetastatic osteosarcoma cell lines. We term these regions metastatic variant enhancer loci (Met-VELs). Met-VELs drive coordinated waves of gene expression during metastatic colonization of the lung. Met-VELs cluster nonrandomly in the genome, indicating that activity of these enhancers and expression of their associated gene targets are positively selected. As evidence of this causal association, osteosarcoma lung metastasis is inhibited by global interruptions of Met-VEL-associated gene expression via pharmacologic BET inhibition, by knockdown of AP-1 transcription factors that occupy Met-VELs, and by knockdown or functional inhibition of individual genes activated by Met-VELs, such as that encoding coagulation factor III/tissue factor (F3). We further show that genetic deletion of a single Met-VEL at the F3 locus blocks metastatic cell outgrowth in the lung. These findings indicate that Met-VELs and the genes they regulate play a functional role in metastasis and may be suitable targets for antimetastatic therapies.


Asunto(s)
Carcinogénesis/genética , Elementos de Facilitación Genéticos/genética , Neoplasias Pulmonares/genética , Osteosarcoma/genética , Línea Celular Tumoral , Epigenómica , Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Metástasis de la Neoplasia/genética , Osteosarcoma/patología , Proteínas/antagonistas & inhibidores , Proteínas/genética , Selección Genética , Tromboplastina/genética , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/genética , Microambiente Tumoral/genética
7.
Nat Commun ; 8: 14400, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28169291

RESUMEN

In addition to mutations in genes, aberrant enhancer element activity at non-coding regions of the genome is a key driver of tumorigenesis. Here, we perform epigenomic enhancer profiling of a cohort of more than forty genetically diverse human colorectal cancer (CRC) specimens. Using normal colonic crypt epithelium as a comparator, we identify enhancers with recurrently gained or lost activity across CRC specimens. Of the enhancers highly recurrently activated in CRC, most are constituents of super enhancers, are occupied by AP-1 and cohesin complex members, and originate from primed chromatin. Many activate known oncogenes, and CRC growth can be mitigated through pharmacologic inhibition or genome editing of these loci. Nearly half of all GWAS CRC risk loci co-localize to recurrently activated enhancers. These findings indicate that the CRC epigenome is defined by highly recurrent epigenetic alterations at enhancers which activate a common, aberrant transcriptional programme critical for CRC growth and survival.


Asunto(s)
Neoplasias Colorrectales/genética , Elementos de Facilitación Genéticos/genética , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica , Sitios Genéticos/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/cirugía , Conjuntos de Datos como Asunto , Epigenómica/métodos , Femenino , Humanos , Ratones , Ratones Desnudos , Mutación , Análisis de Matrices Tisulares , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Neoplasia ; 18(11): 699-710, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27973325

RESUMEN

Metastasis is the cause of more than 90% of all cancer deaths. Despite this fact, most anticancer therapeutics currently in clinical use have limited efficacy in treating established metastases. Here, we identify the endoplasmic reticulum chaperone protein, glucose-regulated protein 78 (GRP78), as a metastatic dependency in several highly metastatic cancer cell models. We find that GRP78 is consistently upregulated when highly metastatic cancer cells colonize the lung microenvironment and that mitigation of GRP78 upregulation via short hairpin RNA or treatment with the small molecule IT-139, which is currently under clinical investigation for the treatment of primary tumors, inhibits metastatic growth in the lung microenvironment. Inhibition of GRP78 upregulation and an associated reduction in metastatic potential have been shown in four highly metastatic cell line models: three human osteosarcomas and one murine mammary adenocarcinoma. Lastly, we show that downmodulation of GRP78 in highly metastatic cancer cells significantly increases median survival times in our in vivo animal model of experimental metastasis. Collectively, our data indicate that GRP78 is an attractive target for the development of antimetastatic therapies.

9.
Clin Cancer Res ; 22(24): 6129-6141, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27342399

RESUMEN

PURPOSE: To successfully metastasize, tumor cells must respond appropriately to biological stressors encountered during metastatic progression. We sought to test the hypothesis that enhanced efficiency of mRNA translation during periods of metastatic stress is required for metastatic competence of osteosarcoma and that this metastasis-specific adaptation is amenable to therapeutic intervention. EXPERIMENTAL DESIGN: We employ novel reporter and proteomic systems that enable tracking of mRNA translation efficiency and output in metastatic osteosarcoma cells as they colonize the lungs. We test the potential to target mRNA translation as an antimetastatic therapeutic strategy through pharmacokinetic studies and preclinical assessment of the prototypic mTOR inhibitor, rapamycin, across multiple models of metastasis. RESULTS: Metastatic osteosarcoma cells translate mRNA more efficiently than nonmetastatic cells during critical stressful periods of metastatic colonization of the lung. Rapamycin inhibits translational output during periods of metastatic stress, mitigates lung colonization, and prolongs survival. mTOR-inhibiting exposures of rapamycin are achievable in mice using treatment schedules that correspond to human doses well below the MTDs defined in human patients, and as such are very likely to be tolerated over long exposures alone and in combination with other agents. CONCLUSIONS: Metastatic competence of osteosarcoma cells is dependent on efficient mRNA translation during stressful periods of metastatic progression, and the mTOR inhibitor, rapamycin, can mitigate this translation and inhibit metastasis in vivo Our data suggest that mTOR pathway inhibitors should be reconsidered in the clinic using rationally designed dosing schedules and clinical metrics related to metastatic progression. Clin Cancer Res; 22(24); 6129-41. ©2016 AACR.


Asunto(s)
Osteosarcoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Osteosarcoma/metabolismo , Fenotipo , Proteómica/métodos , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Crit Rev Oncog ; 20(3-4): 173-97, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26349415

RESUMEN

Osteosarcoma is the most common primary malignancy of bone, typically presenting in the first or second decade of life. Unfortunately, clinical outcomes for osteosarcoma patients have not substantially improved in over 30 years. This stagnation in therapeutic advances is perhaps explained by the genetic, epigenetic, and biological complexities of this rare tumor. In this review we provide a general background on the biology of osteosarcoma and the clinical status quo. We go on to enumerate the genetic and epigenetic defects identified in osteosarcoma. Finally, we discuss ongoing large-scale studies in the field and potential new therapies that are currently under investigation.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/terapia , Epigénesis Genética/genética , Estudios de Asociación Genética/tendencias , Osteosarcoma/genética , Osteosarcoma/terapia , Animales , Neoplasias Óseas/diagnóstico , Estudios de Asociación Genética/métodos , Humanos , MicroARNs/genética , Osteosarcoma/diagnóstico
11.
Cancer Immunol Res ; 3(1): 37-47, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25358764

RESUMEN

Both targeted inhibition of oncogenic driver mutations and immune-based therapies show efficacy in treatment of patients with metastatic cancer, but responses can be either short lived or incompletely effective. Oncogene inhibition can augment the efficacy of immune-based therapy, but mechanisms by which these two interventions might cooperate are incompletely resolved. Using a novel transplantable BRAF(V600E)-mutant murine melanoma model (SB-3123), we explored potential mechanisms of synergy between the selective BRAF(V600E) inhibitor vemurafenib and adoptive cell transfer (ACT)-based immunotherapy. We found that vemurafenib cooperated with ACT to delay melanoma progression without significantly affecting tumor infiltration or effector function of endogenous or adoptively transferred CD8(+) T cells, as previously observed. Instead, we found that the T-cell cytokines IFNγ and TNFα synergized with vemurafenib to induce cell-cycle arrest of tumor cells in vitro. This combinatorial effect was recapitulated in human melanoma-derived cell lines and was restricted to cancers bearing a BRAF(V600E) mutation. Molecular profiling of treated SB-3123 indicated that the provision of vemurafenib promoted the sensitization of SB-3123 to the antiproliferative effects of T-cell effector cytokines. The unexpected finding that immune cytokines synergize with oncogene inhibitors to induce growth arrest has major implications for understanding cancer biology at the intersection of oncogenic and immune signaling and provides a basis for design of combinatorial therapeutic approaches for patients with metastatic cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Inmunoterapia Adoptiva , Indoles/uso terapéutico , Melanoma/terapia , Metástasis de la Neoplasia/terapia , Sulfonamidas/uso terapéutico , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal , Vemurafenib
12.
J Thorac Oncol ; 9(9): 1316-23, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25122427

RESUMEN

BACKGROUND: There is growing interest in defining the somatic mutations associated with small-cell lung cancer (SCLC). Unfortunately, a serious blockade to genomic analyses of this disease is a limited access to tumors because surgery is rarely performed. We used our clinical/pathologic database of SCLC patients to determine the availability of biopsy specimens that could be used for genomic studies and to identify tumors for initial oncogene analysis. METHODS: DNA was extracted from six tumors, three primary and three metastatic, and analyzed by SEQUENOM platform technology. RESULTS: Primary-resected tumor tissue represents less than 3% of all diagnostic specimens in this disease, highlighting the limited access to tissue sufficient for comprehensive genomic analyses. We identified an activating M918T RET somatic mutation in a metastatic SCLC tumor specimen. Bioinformatic search identified RET mutations in other SCLC studies. Stable overexpression of both mutant M918T and wild-type RET in two SCLC cell lines, H1048 and SW1271, activated ERK signaling, MYC expression, and increased cell proliferation, particularly by mutant RET. Stable cells became sensitized to the RET tyrosine kinase inhibitors, vandetanib and ponatinib. Further analysis of RET mRNA expression in SCLC revealed wide variability in both cells and tumors, and SCLC cells demonstrated significantly higher RET expression compared with adenocarcinoma lung cells. CONCLUSIONS: Our data suggest that a subpopulation of SCLC patients may derive benefit from tyrosine kinase inhibitors targeting RET. Coupled with the presence of RET fusion proteins in non-small-cell lung cancer, our data indicate an emerging role for RET in SCLC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Mutación , Proteínas Proto-Oncogénicas c-ret/genética , ARN Neoplásico/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Anciano , Línea Celular Tumoral , Proliferación Celular , Análisis Mutacional de ADN , Estudios de Seguimiento , Humanos , Immunoblotting , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Estudios Retrospectivos , Transducción de Señal , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA