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.
Artículo en Inglés | MEDLINE | ID: mdl-26736815

RESUMEN

Laser ablation (LA) is gaining large acceptance in the treatment of tumor. One of the main risks of this treatment is damaging the healthy tissue around the tumor. Among the solutions proposed to improve the selectivity of the LA and to localize heating to tumor tissue, the use of gold nanoparticles is one of the most promising. The aim of this work is threefold: i) to measure the temperature increase within the tumor during plasmonic photothermal therapy using gold nanorods; ii) to investigate the influence of nanorods concentration and laser settings on both the intra-tumoral temperature and the tumor surface temperature; iii) and to establish the nanorods concentrations able to cause tumor resorption at a defined laser settings. Two sets of trials were performed: i) 16 mice were divided in four groups with different treatment time (i.e., 5 min, 2 min, 1 min, and 30s), with constant gold nanorods amount (i.e., 12.5 µg) and laser power (i.e., 3 W·cm(-2)); ii) 16 mice were divided in four groups treated with different amount of gold nanorods (i.e., control, 12.5 µg, 25 µg, 50 µg) for 5 min at 2 W·cm(-2). Results show significant differences between internal and surface temperatures. We also demonstrate that this temperature difference increases with nanoparticle concentrations, decreases with laser power, and is not influenced by treatment time. This information is critical to improve the theoretical models that will guide future study designs in sensitive orthotopic tumor models.


Asunto(s)
Oro/química , Hipertermia Inducida , Nanotubos/química , Neoplasias/radioterapia , Animales , Línea Celular Tumoral , Humanos , Terapia por Láser , Ratones , Ratones Desnudos , Temperatura , Trasplante Heterólogo
2.
Cancer Gene Ther ; 22(1): 55-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25525033

RESUMEN

In preclinical studies, neural stem cell (NSC)-based delivery of oncolytic virus has shown great promise in the treatment of malignant glioma. Ensuring the success of this therapy will require critical evaluation of the spatial distribution of virus after NSC transplantation. In this study, the patient-derived GBM43 human glioma line was established in the brain of athymic nude mice, followed by the administration of NSCs loaded with conditionally replicating oncolytic adenovirus (NSC-CRAd-S-pk7). We determined the tumor coverage potential of oncolytic adenovirus by examining NSC distribution using magnetic resonance (MR) imaging and by three-dimensional reconstruction from ex vivo tissue specimens. We demonstrate that unmodified NSCs and NSC-CRAd-S-pk7 exhibit a similar distribution pattern with most prominent localization occurring at the tumor margins. We were further able to visualize the accumulation of these cells at tumor sites via T2-weighted MR imaging as well as the spread of viral particles using immunofluorescence. Our analyses reveal that a single administration of oncolytic virus-loaded NSCs allows for up to 31% coverage of intracranial tumors. Such results provide valuable insights into the therapeutic potential of this novel viral delivery platform.


Asunto(s)
Rastreo Celular , Vectores Genéticos/genética , Glioblastoma/genética , Glioblastoma/patología , Imagen por Resonancia Magnética , Células-Madre Neurales/metabolismo , Virus Oncolíticos/genética , Adenoviridae/genética , Animales , Encéfalo/patología , Línea Celular Tumoral , Rastreo Celular/métodos , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Glioblastoma/diagnóstico , Humanos , Ratones , Transducción Genética , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Gene Ther ; 20(12): 678-82, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24310061

RESUMEN

Neural stem cells (NSCs) led to the development of a novel strategy for delivering therapeutic genes to brain tumors. Human NSCs expressing rabbit carboxyl esterase (F3.CE), which activates CPT-11, significantly inhibit the growth of A549 human non-small cell lung adenocarcinoma cells in the presence of CPT-11 in vitro and in vivo. F3.CE cells migrated selectively into the brain metastases located in the opposite hemisphere. The treatment also significantly decreased tumor volume in immune-deficient mice bearing lung cancer when F3.CE cells were transplanted into the contralateral hemisphere. The survival of tumor-bearing animals was significantly prolonged by the treatment with F3.CE and CPT-11. This strategy could be considered as an effective treatment regimen for lung cancer brain metastases.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario , Carboxilesterasa/genética , Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células-Madre Neurales/metabolismo , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Efecto Espectador , Carboxilesterasa/metabolismo , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Conejos , Carga Tumoral/genética
4.
Gene Ther ; 20(2): 143-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22402322

RESUMEN

Medulloblastoma is a heterogeneous diffuse neoplasm that can be highly disseminated, and is the most common malignant childhood brain tumor. Although multimodal treatments have improved survival rates for patients with medulloblastoma, these tumors are associated with high morbidity and mortality. New treatment strategies are urgently needed to improve cure rates and, importantly, to spare normal brain tissue from neurotoxicity and patients from life-long cognitive and functional deficits associated with current therapies. In numerous preclinical brain tumor models, neural stem cells (NSCs) have shown great promise as delivery vehicles for therapeutic genes. Here, we have used an established, genetically modified human NSC line (HB1.F3.CD) to deliver carboxylesterase (CE) to cerebellar tumor foci and locally activate the prodrug camptothecin-11 (CPT-11) (Irinotecan) to the potent topoisomerase I inhibitor SN-38. HB1.F3.CD NSC tumor tropism, intratumoral distribution and therapeutic efficacy were investigated in clinically relevant experimental models. Magnetic resonance imaging was used for in vivo tracking of iron nanoparticle-labeled NSCs, and to assess the therapeutic efficacy of CE-expressing HB1.F3.CD cells. As compared with controls, a significant decrease in tumor growth rate was seen in mice that received both NSCs and CPT-11 as their treatment regimen. Thus, this study provides proof-of-concept for NSC-mediated CE/CPT-11 treatment of medulloblastoma, and serves as a foundation for further studies toward potential clinical application.


Asunto(s)
Carboxilesterasa/genética , Neoplasias Cerebelosas/terapia , Terapia Genética , Meduloblastoma/terapia , Profármacos/uso terapéutico , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Línea Celular Tumoral , Neoplasias Cerebelosas/enzimología , Neoplasias Cerebelosas/genética , Técnicas de Transferencia de Gen , Humanos , Irinotecán , Meduloblastoma/enzimología , Meduloblastoma/genética , Ratones , Ratones Desnudos , Ratones Transgénicos , Células-Madre Neurales/enzimología , Trasplante de Células Madre , Resultado del Tratamiento
5.
Cancer Gene Ther ; 19(6): 431-42, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22555507

RESUMEN

Oncolytic virotherapy is a promising novel therapy for glioblastoma that needs to be optimized before introduced to clinic. The targeting of conditionally replicating adenoviruses (CRAds) can be improved by relying on the tumor-tropic properties of neural stem cells (NSCs). Here, we report the characterization of an FDA approved NSC, HB1.F3-CD, as a cell carrier for CRAd-S-pk7, a glioma-tropic oncolytic adenovirus. We show that NSCs replicate and release infectious CRAd-S-pk7 progeny capable of lysing glioma cell lines. Moreover, ex-vivo-loaded NSCs, injected intracranially in nude mice bearing human glioma xenografts (i) retained their tumor tropism, (ii) continued to replicate CRAd-S-pk7 for more than a week after reaching the tumor site and (iii) successfully handed off CRAd-S-pk7 to glioma cells in vivo. Delivery via carrier cells reduced non-specific adenovirus distribution in the mouse brain. Moreover, we assessed biodistribution of loaded NSCs after intracranial injection in animal models semi-permissive to adenovirus replication, the Syrian hamster and cotton rat. NSCs did not migrate to distant organs and high levels of CRAd-S-pk7 DNA were observed only in the injected hemisphere. In conclusion, this optimized carrier system, with high efficiency of adenovirus delivery and minimal systemic toxicity, poses considerable advantages for anti-glioma oncolytic virotherapy.


Asunto(s)
Adenoviridae/fisiología , Neoplasias Encefálicas/terapia , Glioma/terapia , Células-Madre Neurales/trasplante , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Adenoviridae/genética , Proteínas E1A de Adenovirus/biosíntesis , Proteínas E1A de Adenovirus/genética , Animales , Encéfalo/patología , Encéfalo/virología , Línea Celular , Supervivencia Celular , Cricetinae , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/biosíntesis , Humanos , Luciferasas de Luciérnaga/biosíntesis , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Virus Oncolíticos/genética , Organismos Modificados Genéticamente , Proteínas Recombinantes/biosíntesis , Sigmodontinae , Carga Viral , Replicación Viral
6.
Gene Ther ; 15(10): 739-52, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18369324

RESUMEN

The poor prognosis for patients with aggressive or metastatic tumors and the toxic side effects of currently available treatments necessitate the development of more effective tumor-selective therapies. Stem/progenitor cells display inherent tumor-tropic properties that can be exploited for targeted delivery of anticancer genes to invasive and metastatic tumors. Therapeutic genes that have been inserted into stem cells and delivered to tumors with high selectivity include prodrug-activating enzymes (cytosine deaminase, carboxylesterase, thymidine kinase), interleukins (IL-2, IL-4, IL-12, IL-23), interferon-beta, apoptosis-promoting genes (tumor necrosis factor-related apoptosis-inducing ligand) and metalloproteinases (PEX). We and others have demonstrated that neural and mesenchymal stem cells can deliver therapeutic genes to elicit a significant antitumor response in animal models of intracranial glioma, medulloblastoma, melanoma brain metastasis, disseminated neuroblastoma and breast cancer lung metastasis. Most studies reported reduction in tumor volume (up to 90%) and increased survival of tumor-bearing animals. Complete cures have also been achieved (90% disease-free survival for >1 year of mice bearing disseminated neuroblastoma tumors). As we learn more about the biology of stem cells and the molecular mechanisms that mediate their tumor-tropism and we identify efficacious gene products for specific tumor types, the clinical utility of cell-based delivery strategies becomes increasingly evident.


Asunto(s)
Terapia Genética/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Neoplasias/terapia , Animales , Línea Celular , Técnicas de Transferencia de Gen , Ingeniería Genética , Humanos , Células Madre Mesenquimatosas/fisiología , Células Madre/fisiología
7.
Gene Ther ; 9(10): 613-24, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12032707

RESUMEN

The inherent biology of neural stem cells (NSCs) endows them with capabilities that not only circumvent many of the limitations of other gene transfer vehicles, but that enable a variety of novel therapeutic strategies heretofore regarded as beyond the purview of neural transplantation. Most neurodegenerative diseases are characterized not by discrete, focal abnormalities but rather by extensive, multifocal, or even global neuropathology. Such widely disseminated lesions have not conventionally been regarded as amenable to neural transplantation. However, the ability of NSCs to engraft diffusely and become integral members of structures throughout the host CNS, while also expressing therapeutic molecules, may permit these cells to address that challenge. Intriguingly, while NSCs can be readily engineered to express specified foreign genes, other intrinsic factors appear to emanate spontaneously from NSCs and, in the context of reciprocal donor-host signaling, seem to be capable of neuroprotective and/or neuroregenerative functions. Stem cells additionally have the appealing ability to 'home in' on pathology, even over great distances. Such observations help to advance the idea that NSCs - as a prototype for stem cells from other solid organs - might aid in reconstructing the molecular and cellular milieu of maldeveloped or damaged organs.


Asunto(s)
Sistema Nervioso Central/citología , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Modelos Neurológicos , Regeneración Nerviosa , Traumatismos del Sistema Nervioso/terapia , Adulto , Amiloidosis/terapia , Animales , Isquemia Encefálica/terapia , Neoplasias Encefálicas/terapia , Humanos , Discapacidad Intelectual/terapia , Degeneración Nerviosa/terapia
8.
Proc Natl Acad Sci U S A ; 97(23): 12846-51, 2000 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-11070094

RESUMEN

One of the impediments to the treatment of brain tumors (e.g., gliomas) has been the degree to which they expand, infiltrate surrounding tissue, and migrate widely into normal brain, usually rendering them "elusive" to effective resection, irradiation, chemotherapy, or gene therapy. We demonstrate that neural stem cells (NSCs), when implanted into experimental intracranial gliomas in vivo in adult rodents, distribute themselves quickly and extensively throughout the tumor bed and migrate uniquely in juxtaposition to widely expanding and aggressively advancing tumor cells, while continuing to stably express a foreign gene. The NSCs "surround" the invading tumor border while "chasing down" infiltrating tumor cells. When implanted intracranially at distant sites from the tumor (e.g., into normal tissue, into the contralateral hemisphere, or into the cerebral ventricles), the donor cells migrate through normal tissue targeting the tumor cells (including human glioblastomas). When implanted outside the CNS intravascularly, NSCs will target an intracranial tumor. NSCs can deliver a therapeutically relevant molecule-cytosine deaminase-such that quantifiable reduction in tumor burden results. These data suggest the adjunctive use of inherently migratory NSCs as a delivery vehicle for targeting therapeutic genes and vectors to refractory, migratory, invasive brain tumors. More broadly, they suggest that NSC migration can be extensive, even in the adult brain and along nonstereotypical routes, if pathology (as modeled here by tumor) is present.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/patología , Glioblastoma/patología , Neuronas/fisiología , Nucleósido Desaminasas/genética , Células Madre/fisiología , Animales , Neoplasias Encefálicas/terapia , Movimiento Celular/fisiología , Citosina Desaminasa , Modelos Animales de Enfermedad , Femenino , Terapia Genética/métodos , Glioblastoma/terapia , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Ratones Desnudos , Neuronas/citología , Ratas , Ratas Endogámicas F344 , Células Madre/citología , Tropismo
9.
Mol Ther ; 1(4): 347-57, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10933953

RESUMEN

Cellular delivery of a replication-conditional herpes simplex virus type 1 (HSV-1) vector provides a means for gene therapy of invasive tumor cells. LacZ-bearing neural precursor cells, which can migrate and differentiate in the brain, were infected with a ribonucleotide reductase-deficient HSV-1 mutant virus (rRp450) that replicates only in dividing cells. Replication of rRp450 in neural precursor cells was blocked prior to implantation into the tumor by growth arrest in late G1 phase through treatment with mimosine. Viral titers in the medium of mimosine-treated, rRp450-infected neural precursor cells were below detection levels 3 days after infection. In culture, after removal of mimosine and passaging, cells resumed growth and replication of rRp450 so that, 7 days later, virus was present in the medium and cell death was evident. Mimosine-treated neural precursor cells injected into established intracerebral CNS-1 gliomas in nude mice migrated extensively throughout the tumor and into the surrounding parenchyma beyond the tumor over 3 days. Mimosine-treated neural precursor cells, infected with rRp450 and injected into intracerebral CNS-1 tumors, also migrated within the tumor with the appearance of foci of HSV-thymidine kinase-positive (TK+) cells, presumably including tumor cells, distributed throughout the tumor and in the surrounding parenchyma over a similar period. This migratory cell delivery method has the potential to expand the range of delivery of HSV-1 vectors to tumor cells in the brain.


Asunto(s)
Neoplasias Encefálicas/terapia , Vectores Genéticos , Glioma/terapia , Herpesvirus Humano 1/genética , Neuronas/virología , Células Madre/virología , Animales , Neoplasias Encefálicas/patología , Movimiento Celular , Ganciclovir/farmacología , Genes Virales , Terapia Genética/métodos , Glioma/patología , Proteína Vmw65 de Virus del Herpes Simple/genética , Herpesvirus Humano 1/fisiología , Ratones , Ratones Desnudos , Mimosina/farmacología , Mutación , Neuronas/citología , Neuronas/efectos de los fármacos , Ribonucleótido Reductasas/genética , Células Madre/citología , Células Madre/efectos de los fármacos , Timidina Quinasa/genética , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...