Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Ann Oncol ; 31(12): 1679-1692, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32918998

RESUMEN

Glioblastoma represents the most common primary malignancy of the central nervous system in adults and remains a largely incurable disease. The elucidation of disease subtypes based on mutational profiling, gene expression and DNA methylation has so far failed to translate into improved clinical outcomes. However, new knowledge emerging from the subtyping effort in the IDH-wild-type setting may provide directions for future precision therapies. Here, we review recent learnings in the field, and further consider how tumour microenvironment differences across subtypes may reveal novel contexts of vulnerability. We discuss recent treatment approaches and ongoing trials in the IDH-wild-type glioblastoma setting, and propose an integrated discovery stratagem incorporating multi-omics, single-cell technologies and computational approaches.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Adulto , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Metilación de ADN , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos , Isocitrato Deshidrogenasa/genética , Mutación , Medicina de Precisión , Microambiente Tumoral
2.
Stem Cells Int ; 2016: 5623235, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27274737

RESUMEN

Serum-free culture methods for patient-derived primary glioma cultures, selecting for glioma stem-like cells (GSCs), are becoming the gold standard in neurooncology research. These GSCs can be implemented in drug screens to detect patient-specific responses, potentially bridging the translational gap to personalized medicine. Since numerous compounds are available, a rapid and reliable readout for drug efficacies is required. This can be done using approaches that measure viability, confluency, cytotoxicity, or apoptosis. To determine which assay is best suitable for drug screening, 10 different assays were systematically tested on established glioma cell lines and validated on a panel of GSCs. General applicability was assessed using distinct treatment modalities, being temozolomide, radiation, rapamycin, and the oncolytic adenovirus Delta24-RGD. The apoptosis and cytotoxicity assays did not unequivocally detect responses and were excluded from further testing. The NADH- and ATP-based viability assays revealed comparable readout for all treatments; however, the latter had smaller standard deviations and direct readout. Importantly, drugs that interfere with cell metabolism require alternative techniques such as confluency monitoring to accurately measure treatment effects. Taken together, our data suggest that the combination of ATP luminescence assays with confluency monitoring provides the most specific and reproducible readout for drug screening on primary GSCs.

3.
Gene Ther ; 22(12): 947-59, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26196249

RESUMEN

Oncolytic viruses (OV) have broad potential as an adjuvant for the treatment of solid tumors. The present study addresses the feasibility of clinically applicable drugs to enhance the oncolytic potential of the OV Delta24-RGD in glioblastoma. In total, 446 drugs were screened for their viral sensitizing properties in glioblastoma stem-like cells (GSCs) in vitro. Validation was done for 10 drugs to determine synergy based on the Chou Talalay assay. Mechanistic studies were undertaken to assess viability, replication efficacy, viral infection enhancement and cell death pathway induction in a selected panel of drugs. Four viral sensitizers (fluphenazine, indirubin, lofepramine and ranolazine) were demonstrated to reproducibly synergize with Delta24-RGD in multiple assays. After validation, we underscored general applicability by testing candidate drugs in a broader context of a panel of different GSCs, various solid tumor models and multiple OVs. Overall, this study identified four viral sensitizers, which synergize with Delta24-RGD and two other strains of OVs. The viral sensitizers interact with infection, replication and cell death pathways to enhance efficacy of the OV.


Asunto(s)
Glioblastoma/terapia , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Flufenazina/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/virología , Células HCT116 , Humanos , Indoles/farmacología , Virus Oncolíticos/fisiología , Replicación Viral/efectos de los fármacos
4.
J Neuroimmunol ; 278: 271-6, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25468776

RESUMEN

Inflammation plays a key role in the pathophysiology of Glioblastoma Multiforme (GBM). Here we focus on the contribution of the so far largely ignored complement system. ELISA and immunohistochemistry were combined to assess levels and localization of critical components of the initiation- and effector pathways of the complement cascade in sera and tumor tissue from GBM patients and matched controls. Serum levels of factor-B were decreased in GBM patients whereas C1q levels were increased. C1q and factor-B deposited in the tumor tissue. Deposition of C3 and C5b-9 suggests local complement activation.MBL deficiency, based on serum levels, was significantly less frequent among GBM patients compared to controls (14% vs. 33%). Therefore low levels of MBL may protect against the initiation/progression of GBM.


Asunto(s)
Neoplasias Encefálicas , Activación de Complemento , Glioblastoma , Adulto , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/fisiopatología , Complemento C1q/metabolismo , Factor B del Complemento/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Glioblastoma/sangre , Glioblastoma/inmunología , Glioblastoma/fisiopatología , Humanos , Masculino , Lectina de Unión a Manosa/metabolismo , Persona de Mediana Edad , Análisis de Supervivencia
5.
Cancer Biol Ther ; 7(5): 786-93, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18756624

RESUMEN

Malignant glioma continues to be a major target for gene therapy and virotherapy due to its aggressive growth and the current lack of effective treatment. However, these approaches have been hampered by inefficient infection of glioma cells by viral vectors,particularly vectors derived from serotype 5 adenoviruses (Ad5). This results from limited cell surface expression of the primary adenovirus receptor, coxsackie-adenovirus-receptor (CAR), on tumor cells. To circumvent this problem, Ad fiber pseudotyping,the genetic replacement of either the entire fiber or fiber knob domain with its structural counterpart from another human Ad serotype that recognizes a cellular receptor other than CAR, has been shown to enhance Ad infectivity in a variety of tumor types,including human glioma. Here, we have extended the paradigm of genetic pseudotyping to include fiber domains from non-human or"xenotype" Ads for infectivity enhancement of human glioma cell populations. In this study, we evaluated the gene transfer efficiency of a panel of Ad vectors which express one of five different "xenotype"fiber knob domains, including those derived from murine,ovine, porcine and canine species, in both human glioma cell lines as well as primary glioma tumor cells from patients. Adenovirus vectors displaying either canine Ad or porcine Ad fiber elements had the highest gene transfer to both glioma cell lines and primary tumor cells. The correlation between the viral infectivity of modified adenovirus vectors and expression of human CAR and CD46(an adenovirus type B receptor) on the surfaces of tumor cells was also analyzed. Taken together, human adenovirus vectors modified with "xenotype" fiber elements could be excellent candidates to target human glioma.


Asunto(s)
Adenoviridae/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Animales , Línea Celular Tumoral , Receptor de Androstano Constitutivo , Citomegalovirus/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Humanos , Proteína Cofactora de Membrana/metabolismo , Ratones , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Virus/metabolismo
6.
Oncol Res ; 16(10): 471-7, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18196871

RESUMEN

Multicellular tumor spheroids are used as a model to assess the efficacy of replicating oncolytic adenoviruses. As most assays used to assess cellular viability are unsuitable for oncolytic viruses because of ongoing viral replication, we have used positron emission tomography (PET) to sequentially determine the incorporation of 18F-labeled deoxyglucose (18F-DG) as a measure of viability and compared the results to more commonly used assays for measuring the effect of oncolytic therapy. Glioma monolayer cultures and spheroids were infected with wild-type replicating adenovirus and viability was measured by 18F-DG incorporation, WST-1 assay, crystal violet assay, and spheroid volume 2 to 10 days following infection. Results show that volume measurements in adenovirus-infected spheroids are confounded by the cytopathic effect occurring in infected cells. 18F-DG PET provides a useful method to assess small differences in cell number and viability following oncolytic viral therapy in glioma monolayer cultures and spheroids without the need for disintegration of these cultures. Moreover, using 18F-DG PET, repeated sequential measurements of spheroid viability can be made, decreasing the required number of spheroids per experiment. This is a valuable feature when using spheroids derived from limited amounts of patient material.


Asunto(s)
Adenoviridae/fisiología , Fluorodesoxiglucosa F18 , Glioma/diagnóstico por imagen , Glioma/terapia , Viroterapia Oncolítica/métodos , Glioma/genética , Glioma/virología , Humanos , Tomografía de Emisión de Positrones/métodos , Esferoides Celulares , Células Tumorales Cultivadas
7.
Gene Ther ; 10(23): 1982-91, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14528322

RESUMEN

Conditionally replicative adenoviruses (CRAds) are potentially useful agents for anticancer virotherapy approaches. However, lack of coxsackievirus and adenovirus receptor (CAR) expression on many primary tumor cells limits the oncolytic potency of CRAds. This makes the concept of targeting, that is, redirecting infection via CAR-independent entry pathways, relevant for CRAd development. Bispecific adapter molecules constitute highly versatile means for adenovirus targeting. Here, we constructed a CRAd with the Delta24 E1A mutation that produces a bispecific single-chain antibody directed towards the adenovirus fiber knob and the epidermal growth factor receptor (EGFR). This EGFR-targeted CRAd exhibited increased infection efficiency and oncolytic replication on CAR-deficient cancer cells and augmented lateral spread in CAR-deficient 3-D tumor spheroids in vitro. When compared to its parent control with native tropism, the new CRAd exhibited similar cytotoxicity on CAR-positive cancer cells, but up to 1000-fold enhanced oncolytic potency on CAR-deficient, EGFR-positive cancer cells. In addition, EGFR-targeted CRAd killed primary human CAR-deficient brain tumor specimens that were refractory to the parent control virus. We conclude, therefore, that CRAds expressing bispecific targeting adapter molecules are promising agents for cancer treatment. Their use is likely to result in enhanced oncolytic replication in cancerous tissues and thus in more effective tumor regression.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Receptores ErbB/metabolismo , Terapia Genética/métodos , Vectores Genéticos/genética , Neoplasias/terapia , Receptores Virales/genética , Animales , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Receptores ErbB/inmunología , Marcación de Gen , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores Virales/deficiencia , Replicación Viral
8.
Circ Res ; 91(10): 945-52, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12433840

RESUMEN

Proteases of the plasminogen activator (PA) and matrix metalloproteinase (MMP) system play an important role in smooth muscle cell (SMC) migration and neointima formation after vascular injury. Inhibition of either PAs or MMPs has previously been shown to result in decreased neointima formation in vivo. To inhibit both protease systems simultaneously, a novel hybrid protein, TIMP-1.ATF, was constructed consisting of the tissue inhibitor of metalloproteinase-1 (TIMP-1) domain, as MMP inhibitor, linked to the receptor-binding amino terminal fragment (ATF) of urokinase. By binding to the u-PA receptor this protein will not only anchor the TIMP-1 moiety directly to the cell surface, it will also prevent the local activation of plasminogen by blocking the binding of urokinase-type plasminogen activator (u-PA) to its receptor. Adenoviral expression of TIMP-1.ATF was used to inhibit SMC migration and neointima formation in human saphenous vein segments in vitro. SMC migration was inhibited by 65% in Ad.TIMP-1.ATF-infected cells. Infection with adenoviral vectors encoding the individual domains, Ad.TIMP-1 and Ad.ATF, reduced migration by 32% and 52%, respectively. Neointima formation in saphenous vein organ cultures infected with Ad.TIMP-1.ATF was inhibited by 72% compared with 42% reduction after Ad.TIMP-1 infection and 34% after Ad.ATF infection. These data show that binding of TIMP-1.ATF hybrid protein to the u-PA receptor at the cell surface strongly enhances the inhibitory effect of TIMP-1 on neointima formation in human saphenous vein cultures.


Asunto(s)
Músculo Liso Vascular/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Inhibidor Tisular de Metaloproteinasa-1/genética , Túnica Íntima/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Adenoviridae/genética , Animales , Células CHO/citología , Células CHO/efectos de los fármacos , Células CHO/metabolismo , División Celular/efectos de los fármacos , División Celular/fisiología , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Cricetinae , Medios de Cultivo Condicionados/farmacología , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Técnicas de Transferencia de Gen , Humanos , Técnicas In Vitro , Metaloproteinasa 13 de la Matriz , Inhibidores de la Metaloproteinasa de la Matriz , Músculo Liso Vascular/citología , Estructura Terciaria de Proteína/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Vena Safena/citología , Vena Safena/efectos de los fármacos , Vena Safena/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/farmacología , Túnica Íntima/efectos de los fármacos
9.
Expert Opin Biol Ther ; 2(8): 943-52, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12517272

RESUMEN

Standard therapies are not capable of curing patients with malignant glioma; more than 90% of patients die within 2 years after diagnosis. Gene therapy appeared as a promising new approach for this disease. However, results of clinical trials with replication deficient viral vectors were disappointing. The main reasons being poor transduction efficiency of adenovirus towards glioma cells and limited spread and distribution of the vector in the tumour. With the increasing knowledge of viral genetics and its functions, an attractive alternative tool to kill malignant glioma cells has been developed: Replicating adenovirus as an oncolytic agent. This type of therapy, also referred to as virotherapy, has the potential to overcome some of the limitations connected with replication deficient adenoviral vectors. In this review the authors describe the latest developments in strategies that are being used to create a tumour- or glioma-selective replicating adenovirus. Special attention is given to the methods of viral delivery to an infiltrating tumour in the brain, regarding optimal dose and toxicity. Furthermore, the role of conventional antitumour treatments, such as irradiation and chemotherapy, in enhancing the effect of virotherapy is being emphasised.


Asunto(s)
Adenoviridae , Neoplasias Encefálicas/terapia , Virus , Animales , Línea Celular Tumoral , Glioma/terapia , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA