Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Annu Rev Biochem ; 88: 247-280, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-30901264

RESUMEN

The complexity of human cancer underlies its devastating clinical consequences. Drugs designed to target the genetic alterations that drive cancer have improved the outcome for many patients, but not the majority of them. Here, we review the genomic landscape of cancer, how genomic data can provide much more than a sum of its parts, and the approaches developed to identify and validate genomic alterations with potential therapeutic value. We highlight notable successes and pitfalls in predicting the value of potential therapeutic targets and discuss the use of multi-omic data to better understand cancer dependencies and drug sensitivity. We discuss how integrated approaches to collecting, curating, and sharing these large data sets might improve the identification and prioritization of cancer vulnerabilities as well as patient stratification within clinical trials. Finally, we outline how future approaches might improve the efficiency and speed of translating genomic data into clinically effective therapies and how the use of unbiased genome-wide information can identify novel predictive biomarkers that can be either simple or complex.


Asunto(s)
Genómica , Mutación , Neoplasias/tratamiento farmacológico , Humanos , Neoplasias/genética , Neoplasias/terapia , Medicina de Precisión
2.
Cell ; 162(1): 45-58, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26095253

RESUMEN

Colorectal cancer is a leading cause of cancer-related deaths. Mutations in the innate immune sensor AIM2 are frequently identified in patients with colorectal cancer, but how AIM2 modulates colonic tumorigenesis is unknown. Here, we found that Aim2-deficient mice were hypersusceptible to colonic tumor development. Production of inflammasome-associated cytokines and other inflammatory mediators was largely intact in Aim2-deficient mice; however, intestinal stem cells were prone to uncontrolled proliferation. Aberrant Wnt signaling expanded a population of tumor-initiating stem cells in the absence of AIM2. Susceptibility of Aim2-deficient mice to colorectal tumorigenesis was enhanced by a dysbiotic gut microbiota, which was reduced by reciprocal exchange of gut microbiota with healthy wild-type mice. These findings uncover a synergy between a specific host genetic factor and gut microbiota in determining the susceptibility to colorectal cancer. Therapeutic modulation of AIM2 expression and microbiota has the potential to prevent colorectal cancer.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre/patología , Animales , Azoximetano , Colitis/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Sulfato de Dextran , Enterocitos/patología , Tracto Gastrointestinal/microbiología , Inflamasomas/metabolismo , Ratones , Mutación , Células Madre/metabolismo
3.
Genes Dev ; 36(11-12): 737-751, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35798383

RESUMEN

The primary cilium, a signaling organelle projecting from the surface of a cell, controls cellular physiology and behavior. The presence or absence of primary cilia is a distinctive feature of a given tumor type; however, whether and how the primary cilium contributes to tumorigenesis are unknown for most tumors. Medulloblastoma (MB) is a common pediatric brain cancer comprising four groups: SHH, WNT, group 3 (G3), and group 4 (G4). From 111 cases of MB, we show that primary cilia are abundant in SHH and WNT MBs but rare in G3 and G4 MBs. Using WNT and G3 MB mouse models, we show that primary cilia promote WNT MB by facilitating translation of mRNA encoding ß-catenin, a major oncoprotein driving WNT MB, whereas cilium loss promotes G3 MB by disrupting cell cycle control and destabilizing the genome. Our findings reveal tumor type-specific ciliary functions and underlying molecular mechanisms. Moreover, we expand the function of primary cilia to translation control and reveal a molecular mechanism by which cilia regulate cell cycle progression, thereby providing new frameworks for studying cilium function in normal and pathologic conditions.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Cerebelosas , Meduloblastoma , Animales , Neoplasias Encefálicas/patología , Ciclo Celular/genética , Neoplasias Cerebelosas/genética , Cilios/genética , Humanos , Meduloblastoma/genética , Ratones
4.
Cell ; 157(2): 289-290, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725398

RESUMEN

Attempts to treat cancer with drugs that target mutated proteins have met with mixed success. By screening for compounds that alter the phenotype of glioblastoma cells-an aggressive brain tumor-Kitambi et al. identify a potential new treatment of the disease and shed light on an unusual cell death mechanism.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Piperidinas/farmacología , Quinolinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Humanos
5.
Nature ; 612(7941): 787-794, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36450980

RESUMEN

Medulloblastoma (MB) is the most common malignant childhood brain tumour1,2, yet the origin of the most aggressive subgroup-3 form remains elusive, impeding development of effective targeted treatments. Previous analyses of mouse cerebella3-5 have not fully defined the compositional heterogeneity of MBs. Here we undertook single-cell profiling of freshly isolated human fetal cerebella to establish a reference map delineating hierarchical cellular states in MBs. We identified a unique transitional cerebellar progenitor connecting neural stem cells to neuronal lineages in developing fetal cerebella. Intersectional analysis revealed that the transitional progenitors were enriched in aggressive MB subgroups, including group 3 and metastatic tumours. Single-cell multi-omics revealed underlying regulatory networks in the transitional progenitor populations, including transcriptional determinants HNRNPH1 and SOX11, which are correlated with clinical prognosis in group 3 MBs. Genomic and Hi-C profiling identified de novo long-range chromatin loops juxtaposing HNRNPH1/SOX11-targeted super-enhancers to cis-regulatory elements of MYC, an oncogenic driver for group 3 MBs. Targeting the transitional progenitor regulators inhibited MYC expression and MYC-driven group 3 MB growth. Our integrated single-cell atlases of human fetal cerebella and MBs show potential cell populations predisposed to transformation and regulatory circuitries underlying tumour cell states and oncogenesis, highlighting hitherto unrecognized transitional progenitor intermediates predictive of disease prognosis and potential therapeutic vulnerabilities.


Asunto(s)
Neoplasias Encefálicas , Transformación Celular Neoplásica , Feto , Meduloblastoma , Humanos , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Neoplasias Cerebelosas/patología , Cerebelo/citología , Cerebelo/patología , Feto/citología , Feto/patología , Meduloblastoma/patología , Células-Madre Neurales/citología , Células-Madre Neurales/patología , Pronóstico
6.
Genes Dev ; 34(15-16): 1051-1064, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32675324

RESUMEN

YAP1 is a transcriptional coactivator and the principal effector of the Hippo signaling pathway, which is causally implicated in human cancer. Several YAP1 gene fusions have been identified in various human cancers and identifying the essential components of this family of gene fusions has significant therapeutic value. Here, we show that the YAP1 gene fusions YAP1-MAMLD1, YAP1-FAM118B, YAP1-TFE3, and YAP1-SS18 are oncogenic in mice. Using reporter assays, RNA-seq, ChIP-seq, and loss-of-function mutations, we can show that all of these YAP1 fusion proteins exert TEAD-dependent YAP activity, while some also exert activity of the C'-terminal fusion partner. The YAP activity of the different YAP1 fusions is resistant to negative Hippo pathway regulation due to constitutive nuclear localization and resistance to degradation of the YAP1 fusion proteins. Genetic disruption of the TEAD-binding domain of these oncogenic YAP1 fusions is sufficient to inhibit tumor formation in vivo, while pharmacological inhibition of the YAP1-TEAD interaction inhibits the growth of YAP1 fusion-expressing cell lines in vitro. These results highlight TEAD-dependent YAP activity found in these gene fusions as critical for oncogenesis and implicate these YAP functions as potential therapeutic targets in YAP1 fusion-positive tumors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Ratones , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Señales de Localización Nuclear , Motivos de Nucleótidos , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
7.
Cell ; 145(1): 25-9, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21458665

RESUMEN

Cancer comprises a bewildering assortment of diseases that kill 7.5 million people each year. Poor understanding of cancer's diversity currently thwarts our goal of a cure for every patient, but recent integration of genomic and stem cell technologies promises a route through this impasse.


Asunto(s)
Neoplasias/genética , Animales , Perfilación de la Expresión Génica , Humanos , Leucemia/tratamiento farmacológico , Leucemia/genética , Leucemia/patología , Neoplasias/tratamiento farmacológico , Neoplasias/patología
8.
Nature ; 573(7775): 590-594, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31511697

RESUMEN

The cellular stress response has a vital role in regulating homeostasis by modulating cell survival and death. Stress granules are cytoplasmic compartments that enable cells to survive various stressors. Defects in the assembly and disassembly of stress granules are linked to neurodegenerative diseases, aberrant antiviral responses and cancer1-5. Inflammasomes are multi-protein heteromeric complexes that sense molecular patterns that are associated with damage or intracellular pathogens, and assemble into cytosolic compartments known as ASC specks to facilitate the activation of caspase-1. Activation of inflammasomes induces the secretion of interleukin (IL)-1ß and IL-18 and drives cell fate towards pyroptosis-a form of programmed inflammatory cell death that has major roles in health and disease6-12. Although both stress granules and inflammasomes can be triggered by the sensing of cellular stress, they drive contrasting cell-fate decisions. The crosstalk between stress granules and inflammasomes and how this informs cell fate has not been well-studied. Here we show that the induction of stress granules specifically inhibits NLRP3 inflammasome activation, ASC speck formation and pyroptosis. The stress granule protein DDX3X interacts with NLRP3 to drive inflammasome activation. Assembly of stress granules leads to the sequestration of DDX3X, and thereby the inhibition of NLRP3 inflammasome activation. Stress granules and the NLRP3 inflammasome compete for DDX3X molecules to coordinate the activation of innate responses and subsequent cell-fate decisions under stress conditions. Induction of stress granules or loss of DDX3X in the myeloid compartment leads to a decrease in the production of inflammasome-dependent cytokines in vivo. Our findings suggest that macrophages use the availability of DDX3X to interpret stress signals and choose between pro-survival stress granules and pyroptotic ASC specks. Together, our data demonstrate the role of DDX3X in driving NLRP3 inflammasome and stress granule assembly, and suggest a rheostat-like mechanistic paradigm for regulating live-or-die cell-fate decisions under stress conditions.


Asunto(s)
Muerte Celular/genética , ARN Helicasas DEAD-box/metabolismo , Inflamasomas/genética , Macrófagos/citología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Estrés Fisiológico/genética , Animales , Línea Celular , Supervivencia Celular/genética , ARN Helicasas DEAD-box/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Humanos , Inflamasomas/inmunología , Macrófagos/inmunología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética
9.
Development ; 148(2)2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33472850

RESUMEN

Brain tumours are the commonest solid neoplasms in children, accounting for one quarter of all childhood cancers. Our growing knowledge of basic developmental mechanisms has significantly contributed to understanding the pathogenesis of these tumours and is beginning to impact clinical decisions on how children with these diseases are treated.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/embriología , Encéfalo/patología , Neoplasias Encefálicas/genética , Niño , Humanos , Modelos Biológicos , Microambiente Tumoral
10.
Nature ; 506(7489): 451-5, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24553141

RESUMEN

Members of the nuclear factor-κB (NF-κB) family of transcriptional regulators are central mediators of the cellular inflammatory response. Although constitutive NF-κB signalling is present in most human tumours, mutations in pathway members are rare, complicating efforts to understand and block aberrant NF-κB activity in cancer. Here we show that more than two-thirds of supratentorial ependymomas contain oncogenic fusions between RELA, the principal effector of canonical NF-κB signalling, and an uncharacterized gene, C11orf95. In each case, C11orf95-RELA fusions resulted from chromothripsis involving chromosome 11q13.1. C11orf95-RELA fusion proteins translocated spontaneously to the nucleus to activate NF-κB target genes, and rapidly transformed neural stem cells--the cell of origin of ependymoma--to form these tumours in mice. Our data identify a highly recurrent genetic alteration of RELA in human cancer, and the C11orf95-RELA fusion protein as a potential therapeutic target in supratentorial ependymoma.


Asunto(s)
Transformación Celular Neoplásica , Ependimoma/genética , Ependimoma/metabolismo , FN-kappa B/metabolismo , Proteínas/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Secuencia de Bases , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 11/genética , Ependimoma/patología , Femenino , Humanos , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , FN-kappa B/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas/genética , Factor de Transcripción ReIA/genética , Factores de Transcripción , Translocación Genética/genética , Proteínas Señalizadoras YAP
11.
Lancet Oncol ; 19(6): 768-784, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29778738

RESUMEN

BACKGROUND: Young children with medulloblastoma have a poor overall survival compared with older children, due to use of radiation-sparing therapy in young children. Radiotherapy is omitted or reduced in these young patients to spare them from debilitating long-term side-effects. We aimed to estimate event-free survival and define the molecular characteristics associated with progression-free survival in young patients with medulloblastoma using a risk-stratified treatment strategy designed to defer, reduce, or delay radiation exposure. METHODS: In this multicentre, phase 2 trial, we enrolled children younger than 3 years with newly diagnosed medulloblastoma at six centres in the USA and Australia. Children aged 3-5 years with newly diagnosed, non-metastatic medulloblastoma without any high-risk features were also eligible. Eligible patients were required to start therapy within 31 days from definitive surgery, had a Lansky performance score of at least 30, and did not receive previous radiotherapy or chemotherapy. Patients were stratified postoperatively by clinical and histological criteria into low-risk, intermediate-risk, and high-risk treatment groups. All patients received identical induction chemotherapy (methotrexate, vincristine, cisplatin, and cyclophosphamide), with high-risk patients also receiving an additional five doses of vinblastine. Induction was followed by risk-adapted consolidation therapy: low-risk patients received cyclophosphamide (1500 mg/m2 on day 1), etoposide (100 mg/m2 on days 1 and 2), and carboplatin (area under the curve 5 mg/mL per min on day 2) for two 4-week cycles; intermediate-risk patients received focal radiation therapy (54 Gy with a clinical target volume of 5 mm over 6 weeks) to the tumour bed; and high-risk patients received chemotherapy with targeted intravenous topotecan (area under the curve 120-160 ng-h/mL intravenously on days 1-5) and cyclophosphamide (600 mg/m2 intravenously on days 1-5). After consolidation, all patients received maintenance chemotherapy with cyclophosphamide, topotecan, and erlotinib. The coprimary endpoints were event-free survival and patterns of methylation profiling associated with progression-free survival. Outcome and safety analyses were per protocol (all patients who received at least one dose of induction chemotherapy); biological analyses included all patients with tissue available for methylation profiling. This trial is registered with ClinicalTrials.gov, number NCT00602667, and was closed to accrual on April 19, 2017. FINDINGS: Between Nov 27, 2007, and April 19, 2017, we enrolled 81 patients with histologically confirmed medulloblastoma. Accrual to the low-risk group was suspended after an interim analysis on Dec 2, 2015, when the 1-year event-free survival was estimated to be below the stopping rule boundary. After a median follow-up of 5·5 years (IQR 2·7-7·3), 5-year event-free survival was 31·3% (95% CI 19·3-43·3) for the whole cohort, 55·3% (95% CI 33·3-77·3) in the low-risk cohort (n=23) versus 24·6% (3·6-45·6) in the intermediate-risk cohort (n=32; hazard ratio 2·50, 95% CI 1·19-5·27; p=0·016) and 16·7% (3·4-30·0) in the high-risk cohort (n=26; 3·55, 1·66-7·59; p=0·0011; overall p=0·0021). 5-year progression-free survival by methylation subgroup was 51·1% (95% CI 34·6-67·6) in the sonic hedgehog (SHH) subgroup (n=42), 8·3% (95% CI 0·0-24·0%) in the group 3 subgroup (n=24), and 13·3% (95% CI 0·0-37·6%) in the group 4 subgroup (n=10). Within the SHH subgroup, two distinct methylation subtypes were identified and named iSHH-I and iSHH-II. 5-year progression-free survival was 27·8% (95% CI 9·0-46·6; n=21) for iSHH-I and 75·4% (55·0-95·8; n=21) for iSHH-II. The most common adverse events were grade 3-4 febrile neutropenia (48 patients [59%]), neutropenia (21 [26%]), infection with neutropenia (20 [25%]), leucopenia (15 [19%]), vomiting (15 [19%]), and anorexia (13 [16%]). No treatment-related deaths occurred. INTERPRETATION: The risk-adapted approach did not improve event-free survival in young children with medulloblastoma. However, the methylation subgroup analyses showed that the SHH subgroup had improved progression-free survival compared with the group 3 subgroup. Moreover, within the SHH subgroup, the iSHH-II subtype had improved progression-free survival in the absence of radiation, intraventricular chemotherapy, or high-dose chemotherapy compared with the iSHH-I subtype. These findings support the development of a molecularly driven, risk-adapted, treatment approach in future trials in young children with medulloblastoma. FUNDING: American Lebanese Syrian Associated Charities, St Jude Children's Research Hospital, NCI Cancer Center, Alexander and Margaret Stewart Trust, Sontag Foundation, and American Association for Cancer Research.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/terapia , Irradiación Craneana , Metilación de ADN , Meduloblastoma/genética , Meduloblastoma/terapia , Terapia Neoadyuvante , Factores de Edad , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Australia , Neoplasias Cerebelosas/mortalidad , Neoplasias Cerebelosas/patología , Quimioterapia Adyuvante , Preescolar , Toma de Decisiones Clínicas , Irradiación Craneana/efectos adversos , Irradiación Craneana/mortalidad , Perfilación de la Expresión Génica , Humanos , Lactante , Meduloblastoma/mortalidad , Meduloblastoma/patología , Terapia Neoadyuvante/efectos adversos , Terapia Neoadyuvante/mortalidad , Selección de Paciente , Valor Predictivo de las Pruebas , Supervivencia sin Progresión , Dosis de Radiación , Radioterapia Adyuvante , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Estados Unidos
12.
Nature ; 488(7409): 43-8, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22722829

RESUMEN

Medulloblastoma is a malignant childhood brain tumour comprising four discrete subgroups. Here, to identify mutations that drive medulloblastoma, we sequenced the entire genomes of 37 tumours and matched normal blood. One-hundred and thirty-six genes harbouring somatic mutations in this discovery set were sequenced in an additional 56 medulloblastomas. Recurrent mutations were detected in 41 genes not yet implicated in medulloblastoma; several target distinct components of the epigenetic machinery in different disease subgroups, such as regulators of H3K27 and H3K4 trimethylation in subgroups 3 and 4 (for example, KDM6A and ZMYM3), and CTNNB1-associated chromatin re-modellers in WNT-subgroup tumours (for example, SMARCA4 and CREBBP). Modelling of mutations in mouse lower rhombic lip progenitors that generate WNT-subgroup tumours identified genes that maintain this cell lineage (DDX3X), as well as mutated genes that initiate (CDH1) or cooperate (PIK3CA) in tumorigenesis. These data provide important new insights into the pathogenesis of medulloblastoma subgroups and highlight targets for therapeutic development.


Asunto(s)
Neoplasias Cerebelosas/clasificación , Neoplasias Cerebelosas/genética , Meduloblastoma/clasificación , Meduloblastoma/genética , Mutación/genética , Animales , Antígenos CD , Proteína de Unión a CREB/genética , Cadherinas/genética , Proteínas Cdh1 , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Linaje de la Célula , Neoplasias Cerebelosas/patología , Niño , Fosfatidilinositol 3-Quinasa Clase I , ARN Helicasas DEAD-box/genética , Variaciones en el Número de Copia de ADN , ADN Helicasas/genética , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Genoma Humano/genética , Genómica , Proteínas Hedgehog/metabolismo , Histona Demetilasas/genética , Histonas/metabolismo , Humanos , Meduloblastoma/patología , Metilación , Ratones , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas/genética , Factores de Transcripción/genética , Proteínas Wnt/metabolismo , beta Catenina/genética
13.
Acta Neuropathol ; 133(1): 5-12, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27858204

RESUMEN

Multiple independent genomic profiling efforts have recently identified clinically and molecularly distinct subgroups of ependymoma arising from all three anatomic compartments of the central nervous system (supratentorial brain, posterior fossa, and spinal cord). These advances motivated a consensus meeting to discuss: (1) the utility of current histologic grading criteria, (2) the integration of molecular-based stratification schemes in future clinical trials for patients with ependymoma and (3) current therapy in the context of molecular subgroups. Discussion at the meeting generated a series of consensus statements and recommendations from the attendees, which comment on the prognostic evaluation and treatment decisions of patients with intracranial ependymoma (WHO Grade II/III) based on the knowledge of its molecular subgroups. The major consensus among attendees was reached that treatment decisions for ependymoma (outside of clinical trials) should not be based on grading (II vs III). Supratentorial and posterior fossa ependymomas are distinct diseases, although the impact on therapy is still evolving. Molecular subgrouping should be part of all clinical trials henceforth.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Ependimoma/metabolismo , Ependimoma/terapia , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Consenso , Manejo de la Enfermedad , Ependimoma/genética , Ependimoma/patología , Humanos , Estadificación de Neoplasias
14.
Drug Metab Dispos ; 44(4): 591-4, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26802130

RESUMEN

3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors were potent hits against a mouse ependymoma cell line, but their effectiveness against central nervous system tumors will depend on their ability to cross the blood-brain barrier and attain a sufficient exposure at the tumor. Among 3-hydroxy-3-methylglutaryl coenzyme A inhibitors that had activity in vitro, we prioritized simvastatin (SV) as the lead compound for preclinical pharmacokinetic studies based on its potential for central nervous system penetration as determined from in silico models. Furthermore, we performed systemic plasma disposition and cerebral microdialysis studies of SV (100 mg/kg, p.o.) in a murine model of ependymoma to characterize plasma and tumor extracellular fluid (tECF) pharmacokinetic properties. The murine dosage of SV (100 mg/kg, p.o.) was equivalent to the maximum tolerated dose in patients (7.5 mg/kg, p.o.) based on equivalent plasma exposure of simvastatin acid (SVA) between the two species. SV is rapidly metabolized in murine plasma with 15 times lower exposure compared with human plasma. SVA exposure in tECF was <33.8 ± 11.9 µg/l per hour, whereas the tumor to plasma partition coefficient of SVA was <0.084 ± 0.008. Compared with in vitro washout IC50 values, we did not achieve sufficient exposure of SVA in tECF to suggest tumor growth inhibition; therefore, SV was not carried forward in subsequent preclinical efficacy studies.


Asunto(s)
Neoplasias del Sistema Nervioso Central/metabolismo , Citotoxinas/administración & dosificación , Citotoxinas/metabolismo , Microdiálisis/métodos , Simvastatina/análogos & derivados , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Ratones , Ratones Desnudos , Simvastatina/administración & dosificación , Simvastatina/metabolismo
15.
J Neurooncol ; 126(2): 225-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26518542

RESUMEN

Chemotherapies active in preclinical studies frequently fail in the clinic due to lack of efficacy, which limits progress for rare cancers since only small numbers of patients are available for clinical trials. Thus, a preclinical drug development pipeline was developed to prioritize potentially active regimens for pediatric brain tumors spanning from in vitro drug screening, through intracranial and intra-tumoral pharmacokinetics to in vivo efficacy studies. Here, as an example of the pipeline, data are presented for the combination of 5-fluoro-2'-deoxycytidine and tetrahydrouridine in three pediatric brain tumor models. The in vitro activity of nine novel therapies was tested against tumor spheres derived from faithful mouse models of Group 3 medulloblastoma, ependymoma, and choroid plexus carcinoma. Agents with the greatest in vitro potency were then subjected to a comprehensive series of in vivo pharmacokinetic (PK) and pharmacodynamic (PD) studies culminating in preclinical efficacy trials in mice harboring brain tumors. The nucleoside analog 5-fluoro-2'-deoxycytidine (FdCyd) markedly reduced the proliferation in vitro of all three brain tumor cell types at nanomolar concentrations. Detailed intracranial PK studies confirmed that systemically administered FdCyd exceeded concentrations in brain tumors necessary to inhibit tumor cell proliferation, but no tumor displayed a significant in vivo therapeutic response. Despite promising in vitro activity and in vivo PK properties, FdCyd is unlikely to be an effective treatment of pediatric brain tumors, and therefore was deprioritized for the clinic. Our comprehensive and integrated preclinical drug development pipeline should reduce the attrition of drugs in clinical trials.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Desoxicitidina/análogos & derivados , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Tetrahidrouridina/administración & dosificación , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/sangre , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Relación Dosis-Respuesta a Droga , Epigénesis Genética/efectos de los fármacos , Ratones , Ratones Desnudos , Tetrahidrouridina/sangre , Tetrahidrouridina/farmacocinética , Tetrahidrouridina/uso terapéutico
16.
Nature ; 466(7306): 632-6, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20639864

RESUMEN

Understanding the biology that underlies histologically similar but molecularly distinct subgroups of cancer has proven difficult because their defining genetic alterations are often numerous, and the cellular origins of most cancers remain unknown. We sought to decipher this heterogeneity by integrating matched genetic alterations and candidate cells of origin to generate accurate disease models. First, we identified subgroups of human ependymoma, a form of neural tumour that arises throughout the central nervous system (CNS). Subgroup-specific alterations included amplifications and homozygous deletions of genes not yet implicated in ependymoma. To select cellular compartments most likely to give rise to subgroups of ependymoma, we matched the transcriptomes of human tumours to those of mouse neural stem cells (NSCs), isolated from different regions of the CNS at different developmental stages, with an intact or deleted Ink4a/Arf locus (that encodes Cdkn2a and b). The transcriptome of human supratentorial ependymomas with amplified EPHB2 and deleted INK4A/ARF matched only that of embryonic cerebral Ink4a/Arf(-/-) NSCs. Notably, activation of Ephb2 signalling in these, but not other, NSCs generated the first mouse model of ependymoma, which is highly penetrant and accurately models the histology and transcriptome of one subgroup of human supratentorial tumour. Further, comparative analysis of matched mouse and human tumours revealed selective deregulation in the expression and copy number of genes that control synaptogenesis, pinpointing disruption of this pathway as a critical event in the production of this ependymoma subgroup. Our data demonstrate the power of cross-species genomics to meticulously match subgroup-specific driver mutations with cellular compartments to model and interrogate cancer subgroups.


Asunto(s)
Compartimento Celular , Modelos Animales de Enfermedad , Ependimoma/genética , Ependimoma/patología , Genómica , Mutación/genética , Animales , Sistema Nervioso Central/citología , Sistema Nervioso Central/crecimiento & desarrollo , Neoplasias del Sistema Nervioso Central/clasificación , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/patología , Ependimoma/clasificación , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes p16 , Humanos , Ratones , Modelos Biológicos , Polimorfismo de Nucleótido Simple/genética , Receptor EphB2/genética , Receptor EphB2/metabolismo , Especificidad de la Especie , Células Madre/citología , Células Madre/metabolismo , Sinapsis/metabolismo
17.
Nature ; 468(7327): 1095-9, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21150899

RESUMEN

Medulloblastoma encompasses a collection of clinically and molecularly diverse tumour subtypes that together comprise the most common malignant childhood brain tumour. These tumours are thought to arise within the cerebellum, with approximately 25% originating from granule neuron precursor cells (GNPCs) after aberrant activation of the Sonic Hedgehog pathway (hereafter, SHH subtype). The pathological processes that drive heterogeneity among the other medulloblastoma subtypes are not known, hindering the development of much needed new therapies. Here we provide evidence that a discrete subtype of medulloblastoma that contains activating mutations in the WNT pathway effector CTNNB1 (hereafter, WNT subtype) arises outside the cerebellum from cells of the dorsal brainstem. We found that genes marking human WNT-subtype medulloblastomas are more frequently expressed in the lower rhombic lip (LRL) and embryonic dorsal brainstem than in the upper rhombic lip (URL) and developing cerebellum. Magnetic resonance imaging (MRI) and intra-operative reports showed that human WNT-subtype tumours infiltrate the dorsal brainstem, whereas SHH-subtype tumours are located within the cerebellar hemispheres. Activating mutations in Ctnnb1 had little impact on progenitor cell populations in the cerebellum, but caused the abnormal accumulation of cells on the embryonic dorsal brainstem which included aberrantly proliferating Zic1(+) precursor cells. These lesions persisted in all mutant adult mice; moreover, in 15% of cases in which Tp53 was concurrently deleted, they progressed to form medulloblastomas that recapitulated the anatomy and gene expression profiles of human WNT-subtype medulloblastoma. We provide the first evidence, to our knowledge, that subtypes of medulloblastoma have distinct cellular origins. Our data provide an explanation for the marked molecular and clinical differences between SHH- and WNT-subtype medulloblastomas and have profound implications for future research and treatment of this important childhood cancer.


Asunto(s)
Tronco Encefálico/patología , Neoplasias Cerebelosas/patología , Meduloblastoma/patología , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Mutación , beta Catenina/genética
18.
Development ; 139(18): 3422-31, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22874917

RESUMEN

Neuronal precursors, generated throughout life in the subventricular zone, migrate through the rostral migratory stream to the olfactory bulb where they differentiate into interneurons. We found that the PI3K-Akt-mTorc1 pathway is selectively inactivated in migrating neuroblasts in the subventricular zone and rostral migratory stream, and activated when these cells reach the olfactory bulb. Postnatal deletion of Pten caused aberrant activation of the PI3K-Akt-mTorc1 pathway and an enlarged subventricular zone and rostral migratory stream. This expansion was caused by premature termination of migration and differentiation of neuroblasts and was rescued by inhibition of mTorc1. This phenotype is reminiscent of lamination defects caused by Pten deletion in developing brain that were previously described as defective migration. However, live imaging in acute slices showed that Pten deletion did not cause a uniform defect in the mechanics of directional neuroblast migration. Instead, a subpopulation of Pten-null neuroblasts showed minimal movement and altered morphology associated with differentiation, whereas the remainder showed unimpeded directional migration towards the olfactory bulb. Therefore, migration defects of Pten-null neurons might be secondary to ectopic differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Neuronas/citología , Fosfohidrolasa PTEN/metabolismo , Proteínas/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular/genética , Movimiento Celular/genética , Electroporación , Técnicas In Vitro , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos , Neuronas/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas/genética , Serina-Treonina Quinasas TOR
19.
Development ; 139(10): 1724-33, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22461560

RESUMEN

The WNT pathway plays multiple roles in neural development and is crucial for establishment of the embryonic cerebellum. In addition, WNT pathway mutations are associated with medulloblastoma, the most common malignant brain tumor in children. However, the cell types within the cerebellum that are responsive to WNT signaling remain unknown. Here we investigate the effects of canonical WNT signaling on two important classes of progenitors in the developing cerebellum: multipotent neural stem cells (NSCs) and granule neuron precursors (GNPs). We show that WNT pathway activation in vitro promotes proliferation of NSCs but not GNPs. Moreover, mice that express activated ß-catenin in the cerebellar ventricular zone exhibit increased proliferation of NSCs in that region, whereas expression of the same protein in GNPs impairs proliferation. Although ß-catenin-expressing NSCs proliferate they do not undergo prolonged expansion or neoplastic growth; rather, WNT signaling markedly interferes with their capacity for self-renewal and differentiation. At a molecular level, mutant NSCs exhibit increased expression of c-Myc, which might account for their transient proliferation, but also express high levels of bone morphogenetic proteins and the cyclin-dependent kinase inhibitor p21, which might contribute to their altered self-renewal and differentiation. These studies suggest that the WNT pathway is a potent regulator of cerebellar stem cell growth and differentiation.


Asunto(s)
Cerebelo/citología , Cerebelo/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Cerebelo/embriología , Citometría de Flujo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
20.
Cancer Cell ; 11(1): 69-82, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17222791

RESUMEN

Cancers are believed to arise from cancer stem cells (CSCs), but it is not known if these cells remain dependent upon the niche microenvironments that regulate normal stem cells. We show that endothelial cells interact closely with self-renewing brain tumor cells and secrete factors that maintain these cells in a stem cell-like state. Increasing the number of endothelial cells or blood vessels in orthotopic brain tumor xenografts expanded the fraction of self-renewing cells and accelerated the initiation and growth of tumors. Conversely, depletion of blood vessels from xenografts ablated self-renewing cells from tumors and arrested tumor growth. We propose that brain CSCs are maintained within vascular niches that are important targets for therapeutic approaches.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Células Endoteliales , Células Madre Neoplásicas , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Neoplasias Encefálicas/metabolismo , Comunicación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Femenino , Expresión Génica , Glicoproteínas/metabolismo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Hibridación Fluorescente in Situ , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Neuronas/metabolismo , Neuronas/patología , Péptidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA