Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neuro Oncol ; 25(11): 1932-1946, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37326042

RESUMEN

Glioblastoma (GBM) is the most prevalent malignant primary brain tumor, accounting for 14.2% of all diagnosed tumors and 50.1% of all malignant tumors, and the median survival time is approximately 8 months irrespective of whether a patient receives treatment without significant improvement despite expansive research (Ostrom QT, Price M, Neff C, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2015-2019. Neurooncology. 2022; 24(suppl 5):v1-v95.). Recently, important roles for the circadian clock in GBM tumorigenesis have been reported. Positive regulators of circadian-controlled transcription, brain and muscle ARNT-like 1 (BMAL1), and circadian locomotor output cycles kaput (CLOCK), are highly expressed also in GBM and correlated with poor patient prognosis. BMAL1 and CLOCK promote the maintenance of GBM stem cells (GSCs) and the establishment of a pro-tumorigenic tumor microenvironment (TME), suggesting that targeting the core clock proteins may augment GBM treatment. Here, we review findings that highlight the critical role the circadian clock plays in GBM biology and the strategies by which the circadian clock can be leveraged for GBM treatment in the clinic moving forward.


Asunto(s)
Relojes Circadianos , Glioblastoma , Humanos , Proteínas CLOCK/metabolismo , Factores de Transcripción ARNTL/metabolismo , Encéfalo/metabolismo , Microambiente Tumoral
2.
Int J Health Plann Manage ; 34(1): 241-250, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30109902

RESUMEN

Primary care redesign for older adult patients is currently ongoing in countries with aging populations. One of the main challenges of this type of transformations is how to estimate implementation costs in different types of health care delivery organizations. This study compares start-up and incremental expenses of implementing a primary care redesign across 2 organization types: integrated group (n = 31) practices and independent practice association (IPA) sites (n = 213). Administrators involved with implementing the redesign completed a cost capture template to quantifying expenses. The potential impact of measurement error, recollection bias, and implementation models across sites and geographic regions was examined in sensitivity analyses. Marginal start-up and incremental expenses were higher for Group sites ($122-$328) compared to IPA sites ($31-$227). Group and IPA sites, however, implemented the redesign with different intensities. According to our analyses, if IPA sites implemented the redesign with the same intensity as Group sites, marginal costs would have been $5 to $13 higher for IPA sites than for Group sites. This study shows how a flexible approach to estimate the cost of a wellness care redesign is needed when the intensity of the transformation differs across 2 different types of health care organizations.


Asunto(s)
Organizaciones Responsables por la Atención , Costos y Análisis de Costo/métodos , Práctica de Grupo , Promoción de la Salud/economía , Atención Primaria de Salud/economía , Práctica Privada , Anciano , Prestación Integrada de Atención de Salud , Humanos , Estados Unidos
3.
Sci Transl Med ; 10(443)2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29848664

RESUMEN

Glioblastoma (GBM) is the most lethal primary brain tumor and is highly resistant to current treatments. GBM harbors glioma stem cells (GSCs) that not only initiate and maintain malignant growth but also promote therapeutic resistance including radioresistance. Thus, targeting GSCs is critical for overcoming the resistance to improve GBM treatment. Because the bone marrow and X-linked (BMX) nonreceptor tyrosine kinase is preferentially up-regulated in GSCs relative to nonstem tumor cells and the BMX-mediated activation of the signal transducer and activator of transcription 3 (STAT3) is required for maintaining GSC self-renewal and tumorigenic potential, pharmacological inhibition of BMX may suppress GBM growth and reduce therapeutic resistance. We demonstrate that BMX inhibition by ibrutinib potently disrupts GSCs, suppresses GBM malignant growth, and effectively combines with radiotherapy. Ibrutinib markedly disrupts the BMX-mediated STAT3 activation in GSCs but shows minimal effect on neural progenitor cells (NPCs) lacking BMX expression. Mechanistically, BMX bypasses the suppressor of cytokine signaling 3 (SOCS3)-mediated inhibition of Janus kinase 2 (JAK2), whereas NPCs dampen the JAK2-mediated STAT3 activation via the negative regulation by SOCS3, providing a molecular basis for targeting BMX by ibrutinib to specifically eliminate GSCs while preserving NPCs. Our preclinical data suggest that repurposing ibrutinib for targeting GSCs could effectively control GBM tumor growth both as monotherapy and as adjuvant with conventional therapies.


Asunto(s)
Glioma/patología , Células Madre Neoplásicas/patología , Proteínas Tirosina Quinasas/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Tolerancia a Radiación , Factor de Transcripción STAT3/metabolismo , Adenina/análogos & derivados , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Receptor gp130 de Citocinas/metabolismo , Glioma/terapia , Janus Quinasa 2/metabolismo , Ratones , Modelos Biológicos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Piperidinas , Unión Proteica/efectos de los fármacos , Tolerancia a Radiación/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Análisis de Supervivencia , Temozolomida/farmacología , Temozolomida/uso terapéutico
4.
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
5.
Cancer Res ; 75(8): 1760-9, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25712125

RESUMEN

Glioma stem-like cells (GSC) are a subpopulation of cells in tumors that are believed to mediate self-renewal and relapse in glioblastoma (GBM), the most deadly form of primary brain cancer. In radiation oncology, hyperthermia is known to radiosensitize cells, and it is reemerging as a treatment option for patients with GBM. In this study, we investigated the mechanisms of hyperthermic radiosensitization in GSCs by a phospho-kinase array that revealed the survival kinase AKT as a critical sensitization determinant. GSCs treated with radiation alone exhibited increased AKT activation, but the addition of hyperthermia before radiotherapy reduced AKT activation and impaired GSC proliferation. Introduction of constitutively active AKT in GSCs compromised hyperthermic radiosensitization. Pharmacologic inhibition of PI3K further enhanced the radiosensitizing effects of hyperthermia. In a preclinical orthotopic transplant model of human GBM, thermoradiotherapy reduced pS6 levels, delayed tumor growth, and extended animal survival. Together, our results offer a preclinical proof-of-concept for further evaluation of combined hyperthermia and radiation for GBM treatment.


Asunto(s)
Glioma/terapia , Hipertermia Inducida , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Proteína Oncogénica v-akt/antagonistas & inhibidores , Tolerancia a Radiación , Animales , Muerte Celular/efectos de la radiación , Proliferación Celular/genética , Proliferación Celular/efectos de la radiación , Células Cultivadas , Terapia Combinada , Reparación del ADN/efectos de la radiación , Glioma/genética , Glioma/patología , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/patología , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/metabolismo , Tolerancia a Radiación/genética , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación
6.
Drugs Today (Barc) ; 42(10): 657-70, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17136225

RESUMEN

In clinical trials thus far, single-targeted kinase inhibitors have shown only limited success in demonstrating survival benefits in cancer. This has led to the development of multitargeted kinase inhibitors capable of disrupting various mitogenic pathways in both cancer cells and associated vasculature. Vandetanib is a novel multitargeted kinase inhibitor exhibiting potent activity against vascular endothelial growth factor receptor-2 (VEGFR-2; kinase insert domain-containing receptor [KDR]) and, to a lesser extent, epidermal growth factor receptor (EGFR) and RET kinase. Vascular endothelial growth factor (VEGF) and VEGFR-2 play a pivotal role in regulating angiogenesis and vascular permeability in cancers. In addition to its antiangiogenic effects, vandetanib acts against EGFR, which is overexpressed or mutated in several solid tumors. Furthermore, vandetanib exerts activity against oncogenic RET kinase, the overexpression of which is common in medullary and papillary thyroid carcinomas. Therefore, the multitargeted kinase inhibitor vandetanib represents a new approach, targeting both tumor cells and tumor-associated endothelial cells. Preclinical studies of vandetanib have demonstrated antitumor efficacy against multiple human cancer xenografts in subcutaneous, orthotopic and metastatic models. Phase I clinical trials have demonstrated that vandetanib is well tolerated. Common adverse events included rash, diarrhea and asymptomatic QTc prolongation. Phase II clinical studies in patients with non-small-cell lung cancer have shown promising results, employing vandetanib as both monotherapy and in combination with docetaxel. Phase II studies in other cancers have likewise been initiated. This review summarizes preclinical and clinical studies of vandetanib for the treatment of cancers.


Asunto(s)
Neoplasias/tratamiento farmacológico , Piperidinas/uso terapéutico , Quinazolinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos/métodos , Humanos , Estructura Molecular , Piperidinas/química , Piperidinas/farmacología , Quinazolinas/química , Quinazolinas/farmacología , Resultado del Tratamiento
7.
J Neurooncol ; 65(1): 27-35, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14649883

RESUMEN

TP-38 is a recombinant chimeric targeted toxin composed of the EGFR binding ligand TGF-alpha and a genetically engineered form of the Pseudomonas exotoxin, PE-38. After in vitro and in vivo animal studies that showed specific activity and defined the maximum tolerated dose (MTD), we investigated this agent in a Phase I trial. The primary objective of this study was to define the MTD and dose limiting toxicity of TP-38 delivered by convection-enhanced delivery in patients with recurrent malignant brain tumors. Twenty patients were enrolled in the study and doses were escalated from 25 ng/mL to 100 with a 40 mL infusion volume delivered by two catheters. One patient developed Grade IV fatigue at the 100 ng/mL dose, but the MTD has not been established. The overall median survival after TP-38 for all patients was 23 weeks whereas for those without radiographic evidence of residual disease at the time of therapy, the median survival was 31.9 weeks. Overall, 3 of 15 patients, with residual disease at the time of therapy, have demonstrated radiographic responses and one patient with a complete response and has survived greater than 83 weeks.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Exotoxinas/administración & dosificación , Glioblastoma/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Factor de Crecimiento Transformador alfa/administración & dosificación , Adulto , Anciano , Neoplasias Encefálicas/mortalidad , Evaluación Preclínica de Medicamentos , Femenino , Glioblastoma/mortalidad , Humanos , Infusiones Parenterales , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Pseudomonas aeruginosa/química , Tasa de Supervivencia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA