Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 14(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36497300

RESUMEN

Therapeutic efficacy of retroviral replicating vector (RRV)-mediated prodrug activator gene therapy has been demonstrated in a variety of tumor models, but clinical investigation of this approach has so far been restricted to glioma and gastrointestinal malignancies. In the present study, we evaluated replication kinetics, transduction efficiency, and therapeutic efficacy of RRV in experimental models of lung cancer. RRV delivering GFP as a reporter gene showed rapid viral replication in a panel of lung cancer cells in vitro, as well as robust intratumoral replication and high levels of tumor transduction in subcutaneous and orthotopic pleural dissemination models of lung cancer in vivo. Toca 511 (vocimagene amiretrorepvec), a clinical-stage RRV encoding optimized yeast cytosine deaminase (yCD) which converts the prodrug 5-fluorocytosine (5-FC) to the active drug 5-fluorouracil (5-FU), showed potent cytotoxicity in lung cancer cells upon exposure to 5-FC prodrug. In vivo, Toca 511 achieved significant tumor growth inhibition following 5-FC treatment in subcutaneous and orthotopic pleural dissemination models of lung cancer in both immunodeficient and immunocompetent hosts, resulting in significantly increased overall survival. This study demonstrates that RRV can serve as highly efficient vehicles for gene delivery to lung cancer, and indicates the translational potential of RRV-mediated prodrug activator gene therapy with Toca 511/5-FC as a novel therapeutic strategy for pulmonary malignancies.

2.
Cancer Gene Ther ; 25(7-8): 184-195, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29735994

RESUMEN

Toca 511, a retroviral replicating vector (RRV) encoding the yeast cytosine deaminase (yCD) prodrug activator gene, which mediates conversion of the prodrug 5-fluorocytosine (5-FC) to the anticancer drug 5-fluorouracil (5-FU), is currently being evaluated in Phase II/III clinical trials for glioma, and showing highly promising evidence of therapeutic activity. Here we evaluated RRV-mediated prodrug activator gene therapy as a new therapeutic approach for pancreatic ductal adenocarcinoma (PDAC). RRV spread rapidly and conferred significant cytotoxicity with prodrug in a panel of PDAC cells. Efficient intratumoral replication and complete inhibition of tumor growth upon 5-FC administration were observed in both immunodeficient and immunocompetent subcutaneous PDAC models. Biodistribution of RRV was highly restricted in normal tissues, especially in immunocompetent hosts. Tumor growth inhibition by Toca 511 followed by 5-FC was also confirmed in the orthotopic PDAC model. This study provides the first proof-of-concept for application of Toca 511 and Toca FC (extended release 5-FC) to the treatment of human PDAC, and provided support for inclusion of PDAC in a Phase I study evaluating Toca 511 in various systemic malignancies, (NCT02576665), which has recently been initiated.


Asunto(s)
Citosina Desaminasa , Fluorouracilo/farmacología , Terapia Genética/métodos , Vectores Genéticos , Neoplasias Pancreáticas , Profármacos/farmacología , Retroviridae , Proteínas de Saccharomyces cerevisiae , Línea Celular Tumoral , Citosina Desaminasa/biosíntesis , Citosina Desaminasa/genética , Fluorouracilo/farmacocinética , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Profármacos/farmacocinética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/biosíntesis , Proteínas de Saccharomyces cerevisiae/genética
3.
Neuro Oncol ; 19(7): 918-929, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28387831

RESUMEN

BACKGROUND: Prodrug-activator gene therapy with Toca 511, a tumor-selective retroviral replicating vector (RRV) encoding yeast cytosine deaminase, is being evaluated in recurrent high-grade glioma patients. Nonlytic retroviral infection leads to permanent integration of RRV into the cancer cell genome, converting infected cancer cell and progeny into stable vector producer cells, enabling ongoing transduction and viral persistence within tumors. Cytosine deaminase in infected tumor cells converts the antifungal prodrug 5-fluorocytosine into the anticancer drug 5-fluorouracil, mediating local tumor destruction without significant systemic adverse effects. METHODS: Here we investigated mechanisms underlying the therapeutic efficacy of this approach in orthotopic brain tumor models, employing both human glioma xenografts in immunodeficient hosts and syngeneic murine gliomas in immunocompetent hosts. RESULTS: In both models, a single injection of replicating vector followed by prodrug administration achieved long-term survival benefit. In the immunodeficient model, tumors recurred repeatedly, but bioluminescence imaging of tumors enabled tailored scheduling of multicycle prodrug administration, continued control of disease burden, and long-term survival. In the immunocompetent model, complete loss of tumor signal was observed after only 1-2 cycles of prodrug, followed by long-term survival without recurrence for >300 days despite discontinuation of prodrug. Long-term survivors rejected challenge with uninfected glioma cells, indicating immunological responses against native tumor antigens, and immune cell depletion showed a critical role for CD4+ T cells. CONCLUSION: These results support dual mechanisms of action contributing to the efficacy of RRV-mediated prodrug-activator gene therapy: long-term tumor control by prodrug conversion-mediated cytoreduction, and induction of antitumor immunity.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/inmunología , Glioma/terapia , Recurrencia Local de Neoplasia/terapia , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular , Citosina Desaminasa/genética , Femenino , Vectores Genéticos/fisiología , Glioma/patología , Humanos , Ratones , Retroviridae/fisiología , Análisis de Supervivencia
4.
Neuro Oncol ; 19(7): 930-939, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28387849

RESUMEN

BACKGROUND: Toca 511 (vocimagene amiretrorepvec) is a retroviral replicating vector encoding an optimized yeast cytosine deaminase (CD). Tumor-selective expression of CD converts the prodrug, 5-fluorocytosine (5-FC), into the active chemotherapeutic, 5-fluorouracil (5-FU). This therapeutic approach is being tested in a randomized phase II/III trial in recurrent glioblastoma and anaplastic astrocytoma (NCT0241416). The aim of this study was to identify the immune cell subsets contributing to antitumor immune responses following treatment with 5-FC in Toca 511-expressing gliomas in a syngeneic mouse model. METHODS: Flow cytometry was utilized to monitor and characterize the immune cell infiltrate in subcutaneous Tu-2449 gliomas in B6C3F1 mice treated with Toca 511 and 5-FC. RESULTS: Tumor-bearing animals treated with Toca 511 and 5-FC display alterations in immune cell populations within the tumor that result in antitumor immune protection. Attenuated immune subsets were exclusive to immunosuppressive cells of myeloid origin. Depletion of immunosuppressive cells temporally preceded a second event which included expansion of T cells which were polarized away from Th2 and Th17 in the CD4+ T cell compartment with concomitant expansion of interferon gamma-expressing CD8+ T cells. Immune alterations correlated with clearance of Tu-2449 subcutaneous tumors and T cell-dependent protection from future tumor challenge. CONCLUSIONS: Treatment with Toca 511 and 5-FC has a concentrated effect at the site of the tumor which causes direct tumor cell death and alterations in immune cell infiltrate, resulting in a tumor microenvironment that is more permissive to establishment of a T cell mediated antitumor immune response.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Flucitosina/uso terapéutico , Glioma/tratamiento farmacológico , Glioma/inmunología , Animales , Línea Celular Tumoral , Citosina Desaminasa , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Inmunidad , Ratones , Monocitos/efectos de los fármacos , Células Mieloides/efectos de los fármacos , Profármacos/uso terapéutico , Retroviridae , Linfocitos T/efectos de los fármacos
5.
Cancer Immunol Immunother ; 65(9): 1085-97, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27439500

RESUMEN

Natural killer (NK) cells are functionally suppressed in the glioblastoma multiforme (GBM) tumor microenvironment. We have recently shown that survival and differentiation of cancer stem-like cells (CSCs)/poorly differentiated tumors are controlled through two distinct phenotypes of cytotoxic and non-cytotoxic/split anergized NK cells, respectively. In this paper, we studied the function of NK cells against brain CSCs/poorly differentiated GBM and their NK cell-differentiated counterparts. Brain CSCs/poorly differentiated GBM, differentiated by split anergized NK supernatants (supernatants from NK cells treated with IL-2 + anti-CD16mAb) expressed higher levels of CD54, B7H1 and MHC-I and were killed less by the NK cells, whereas their CSCs/poorly differentiated counterparts were highly susceptible to NK cell lysis. Resistance to NK cells and differentiation of brain CSCs/poorly differentiated GBM by split anergized NK cells were mediated by interferon (IFN)-γ and tumor necrosis factor (TNF)-α. Brain CSCs/poorly differentiated GBM expressed low levels of TNFRs and IFN-γRs, and when differentiated and cultured with IL-2-treated NK cells, they induced increased secretion of pro-inflammatory cytokine interleukin (IL)-6 and chemokine IL-8 in the presence of decreased IFN-γ secretion. NK-induced differentiation of brain CSCs/poorly differentiated GBM cells was independent of the function of IL-6 and/or IL-8. The inability of NK cells to lyse GBM tumors and the presence of a sustained release of pro-inflammatory cytokines IL-6 and chemokine IL-8 in the presence of a decreased IFN-γ secretion may lead to the inadequacy of NK cells to differentiate GBM CSCs/poorly differentiated tumors, thus failing to control tumor growth.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Interferón gamma/inmunología , Interleucina-6/inmunología , Interleucina-8/inmunología , Células Asesinas Naturales/inmunología , Células Madre Neoplásicas/inmunología , Neoplasias Encefálicas/patología , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Glioblastoma/patología , Humanos , Interferón gamma/deficiencia , Interleucina-2/farmacología , Células Madre Neoplásicas/patología
6.
J Cancer ; 6(9): 866-76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26284138

RESUMEN

We have recently shown that Natural Killer (NK) cells control survival and differentiation of Cancer Stem-like Cells (CSCs) through two distinct phenotypes of cytotoxic and anergic NK cells, respectively. In this report, brain CSCs and their serum and NK cell differentiated counterparts were studied. Serum-differentiated brain CSCs were significantly less susceptible to NK cells and CTL direct cytotoxicity as well as NK cell mediated Antibody Dependent Cellular Cytotoxicity (ADCC), whereas their CSCs were highly susceptible. The levels of CD44 and EGFR were higher in brain tumor CSCs when compared to the serum-differentiated tumors. No differences could be observed for the expression of MHC class I between brain tumor stem cells and their serum-differentiated counterparts. Moreover, supernatants from the combination of IL-2 and anti-CD16mAb treated NK cells (anergized NK cells) induced resistance of brain tumor CSCs to NK cell mediated cytotoxicity. Unlike serum-differentiated CSCs, NK supernatant induced differentiation and resistance to cytotoxicity in brain CSCs correlated with the increased expression of CD54 and MHC class I. The addition of anti-MHC class I antibody moderately inhibited NK mediated cytotoxicity against untreated or serum-differentiated CSCs, whereas it increased cytotoxicity against NK supernatant differentiated tumors. Therefore, two distinct mechanisms govern serum and NK supernatant mediated differentiation of brain tumors.

7.
Cancer Gene Ther ; 21(10): 405-410, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25301172

RESUMEN

A tumor-selective non-lytic retroviral replicating vector (RRV), Toca 511, and an extended-release formulation of 5-fluorocytosine (5-FC), Toca FC, are currently being evaluated in clinical trials in patients with recurrent high-grade glioma (NCT01156584, NCT01470794 and NCT01985256). Tumor-selective propagation of this RRV enables highly efficient transduction of glioma cells with cytosine deaminase (CD), which serves as a prodrug activator for conversion of the anti-fungal prodrug 5-FC to the anti-cancer drug 5-fluorouracil (5-FU) directly within the infected cells. We investigated whether, in addition to its direct cytotoxic effects, 5-FU generated intracellularly by RRV-mediated CD/5-FC prodrug activator gene therapy could also act as a radiosensitizing agent. Efficient transduction by RRV and expression of CD were confirmed in the highly aggressive, radioresistant human glioblastoma cell line U87EGFRvIII and its parental cell line U87MG (U87). RRV-transduced cells showed significant radiosensitization even after transient exposure to 5-FC. This was confirmed both in vitro by a clonogenic colony survival assay and in vivo by bioluminescence imaging analysis. These results provide a convincing rationale for development of tumor-targeted radiosensitization strategies utilizing the tumor-selective replicative capability of RRV, and incorporation of radiation therapy into future clinical trials evaluating Toca 511 and Toca FC in brain tumor patients.


Asunto(s)
Fluorouracilo/metabolismo , Vectores Genéticos/genética , Glioma/genética , Glioma/metabolismo , Profármacos/metabolismo , Tolerancia a Radiación/genética , Retroviridae/genética , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Femenino , Fluorouracilo/farmacología , Expresión Génica , Orden Génico , Técnicas de Transferencia de Gen , Genes Reporteros , Genes Transgénicos Suicidas , Terapia Genética , Glioma/patología , Glioma/terapia , Humanos , Ratones , Profármacos/farmacología , Transducción Genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Biol Chem ; 289(20): 13974-85, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24644285

RESUMEN

Despite recent advances in molecular classification, surgery, radiotherapy, and targeted therapies, the clinical outcome of patients with malignant brain tumors remains extremely poor. In this study, we have identified the tetraspan protein epithelial membrane protein-2 (EMP2) as a potential target for glioblastoma (GBM) killing. EMP2 had low or undetectable expression in normal brain but was highly expressed in GBM as 95% of patients showed some expression of the protein. In GBM cells, EMP2 enhanced tumor growth in vivo in part by up-regulating αvß3 integrin surface expression, activating focal adhesion kinase and Src kinases, and promoting cell migration and invasion. Consistent with these findings, EMP2 expression significantly correlated with activated Src kinase in patient samples and promoted tumor cell invasion using intracranial mouse models. As a proof of principle to determine whether EMP2 could serve as a target for therapy, cells were treated using specific anti-EMP2 antibody reagents. These reagents were effective in killing GBM cells in vitro and in reducing tumor load in subcutaneous mouse models. These results support the role of EMP2 in the pathogenesis of GBM and suggest that anti-EMP2 treatment may be a novel therapeutic treatment.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Glicoproteínas de Membrana/metabolismo , Terapia Molecular Dirigida , Familia-src Quinasas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Activación Enzimática , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/enzimología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Glicoproteínas de Membrana/inmunología , Ratones , Fenotipo
9.
Clin Cancer Res ; 19(15): 4137-48, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23780889

RESUMEN

PURPOSE: Individual or combined strategies of cellular therapy with alloreactive CTLs (alloCTL) and gene therapy using retroviral replicating vectors (RRV) encoding a suicide prodrug activating gene were explored for the treatment of breast tumors metastatic to the brain. EXPERIMENTAL DESIGN: AlloCTL, sensitized to the HLA of MDA-MB-231 breast cancer cells, were examined in vitro for antitumor functionality toward breast cancer targets. RRV encoding the yeast cytosine deaminase (CD) gene was tested in vivo for virus spread, ability to infect, and kill breast cancer targets when exposed to 5-fluorocytosine (5-FC). Individual and combination treatments were tested in subcutaneous and intracranial xenograft models with 231BR, a brain tropic variant. RESULTS: AlloCTL preparations were cytotoxic, proliferated, and produced IFN-γ when coincubated with target cells displaying relevant HLA. In vivo, intratumorally placed alloCTL trafficked through one established intracranial 231BR focus to another in contralateral brain and induced tumor cell apoptosis. RRV-CD efficiently spread in vivo, infected 231BR and induced their apoptosis upon 5-FC exposure. Subcutaneous tumor volumes were significantly reduced in alloCTL and/or gene therapy-treated groups compared to control groups. Mice with established intracranial 231BR tumors treated with combined alloCTL and RRV-CD had a median survival of 97.5 days compared with single modalities (50-83 days); all experimental treatment groups survived significantly longer than sham-treated groups (median survivals 31.5 or 40 days) and exhibited good safety/toxicity profiles. CONCLUSION: The results indicate combining cellular and suicide gene therapies is a viable strategy for the treatment of established breast tumors in the brain.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Terapia Genética , Linfocitos T Citotóxicos , Adenoviridae , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Terapia Combinada , Citosina Desaminasa/genética , Citosina Desaminasa/uso terapéutico , Femenino , Flucitosina/administración & dosificación , Genes Transgénicos Suicidas/genética , Vectores Genéticos , Humanos , Ratones , Profármacos/administración & dosificación
10.
Mol Ther ; 20(9): 1689-98, 2012 09.
Artículo en Inglés | MEDLINE | ID: mdl-22547150

RESUMEN

Retroviral replicating vectors (RRVs) are a nonlytic alternative to oncolytic replicating viruses as anticancer agents, being selective both for dividing cells and for cells that have defects in innate immunity and interferon responsiveness. Tumor cells fit both these descriptions. Previous publications have described a prototype based on an amphotropic murine leukemia virus (MLV), encoding yeast cytosine deaminase (CD) that converts the prodrug 5-fluorocytosine (5-FC) to the potent anticancer drug, 5-fluorouracil (5-FU) in an infected tumor. We report here the selection of one lead clinical candidate based on a general design goal to optimize the genetic stability of the virus and the CD activity produced by the delivered transgene. Vectors were tested for titer, genetic stability, CD protein and enzyme activity, ability to confer susceptibility to 5-FC, and preliminary in vivo antitumor activity and stability. One vector, Toca 511, (aka T5.0002) encoding an optimized CD, shows a threefold increased specific activity in infected cells over infection with the prototype RRV and shows markedly higher genetic stability. Animal testing demonstrated that Toca 511 replicates stably in human tumor xenografts and, after 5-FC administration, causes complete regression of such xenografts. Toca 511 (vocimagene amiretrorepvec) has been taken forward to preclinical and clinical trials.


Asunto(s)
Terapia Genética/métodos , Virus de la Leucemia Murina/genética , Neoplasias Experimentales/terapia , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Flucitosina/metabolismo , Flucitosina/farmacología , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Expresión Génica , Vectores Genéticos , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Profármacos/metabolismo , Profármacos/farmacología , Estabilidad del ARN , Ratas , Transgenes
11.
Curr Gene Ther ; 12(1): 20-32, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22263921

RESUMEN

As the interest in gene therapy increases, the development of an efficient and reliable means to monitor gene delivery and expression in patients is becoming more important. An ideal imaging modality would be non-invasive, allowing for repeated imaging, thus validating stages subsequent to vector administration and allowing for the improvement of clinical protocols. Positron Emission Tomography (PET) has been employed for some time in clinical imaging and has in more recent years been adapted to enable imaging in small animal models, including gene therapy models for a range of diseases. PET imaging is based on the detection of trace quantities of positron-emitting molecular probe within cells postadministration, permitting imaging of target molecules in vivo, and numerous tracers have been developed for a wide range of applications, including imaging of reporter gene activity. Use of radiolabelled substrates that interact with specific transgene proteins, has identified a number of reporter genes that are suitable for imaging vector mediated gene delivery and expression in both pre-clinical and clinical situations. These reporter genes enable non-invasive analysis of the location, level and kinetics of transgene activity. Among the various imaging modalities in existence, the PET approach displays arguably the optimum characteristics in terms of sensitivity and quantitation for in vivo gene expression measurements. Given the existing availability of PET scanning equipment and expertise in hospitals, this imaging modality represents the most clinically applicable means of analysing gene therapy in patients. This review outlines the principles of PET imaging in the context of gene and cell therapy at both pre-clinical and clinical levels, comparing PET with other relevant modalities, and describes the progress to date in this field.


Asunto(s)
Expresión Génica , Genes Reporteros , Tomografía de Emisión de Positrones/métodos , Timidina Quinasa/genética , Transgenes , Animales , Rastreo Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Terapia Genética/métodos , Humanos , Ratones , Timidina Quinasa/análisis
12.
J Biol Chem ; 283(16): 10958-66, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18292095

RESUMEN

A cancer stem cell population in malignant brain tumors takes an essential part in brain tumor initiation, growth, and recurrence. Growth factors, such as epidermal growth factor, fibroblast growth factor-2, vascular endothelial growth factor, platelet-derived growth factor, and hepatocyte growth factor, are shown to support the proliferation of neural stem cells and also may play key roles in gliomagenesis. However, the responsible growth factor(s), which controls maintenance of brain tumor stem cells, is not yet uncovered. We have established three cancer stem cell lines from human gliomas. These cells were immunoreactive with the neuronal progenitor markers, nestin and CD133, and established tumors that closely resembled the features of original tumor upon transplantation into mouse brain. Three cell lines retained their self-renewal ability and proliferation only in the presence of epidermal growth factor (>2.5 ng/ml). In sharp contrast, other growth factors, including fibroblast growth factor-2, failed to support maintenance of these cells. The tyrosine kinase inhibitors of epidermal growth factor signaling (AG1478 and gefitinib) suppressed the proliferation and self-renewal of these cells. Gefitinib inhibited phosphorylation of epidermal growth factor receptor as well as Akt kinase and extracellular signal-regulated kinase 1/2. Flow cytometric analysis revealed that epidermal growth factor concentration-dependently increased the population of CD133-positive cells. Gefitinib significantly reduced CD133-positive fractions and also induced their apoptosis. These results indicate that maintenance of human brain tumor stem cells absolutely requires epidermal growth factor and that tyrosine kinase inhibitors of epidermal growth factor signaling potentially inhibit proliferation and induce apoptosis of these cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Factor de Crecimiento Epidérmico/fisiología , Regulación Neoplásica de la Expresión Génica , Células Madre/metabolismo , Antígeno AC133 , Antígenos CD/biosíntesis , Inhibidores Enzimáticos/farmacología , Gefitinib , Glicoproteínas/biosíntesis , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Biológicos , Péptidos , Fosforilación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/farmacología , Transducción de Señal , Células Tumorales Cultivadas , Tirfostinos/farmacología
13.
Biochem Biophys Res Commun ; 361(3): 586-92, 2007 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-17673180

RESUMEN

There is increasing evidence for the presence of cancer stem-like progenitors in malignant brain tumors. This subpopulation of progenitor cells, the so-called "cancer stem cells (CSCs)", may play a pivotal role in brain tumor initiation, growth, and recurrence. Here we describe the establishment of one permanent brain tumor stem cell line that able to form new spheres after culture under adherent monolayer conditions and to recapitulate the properties of the original tumor upon transplantation into immunodeficient mice. Re-formed spheres retained their stem cell properties and isolated single CSCs from these spheres formed spheres/tumors even after long-term cultures (over 2 years). These data suggested that a small population of CSCs preserved its stem cell properties even after serial passages under non-adherent/adherent culture conditions. Evaluation of underlying metabolic events and assessment of the biological features of these viable cell lines will yield useful knowledge on the in situ behavior of brain tumors.


Asunto(s)
Neoplasias Encefálicas/patología , Células Madre Neoplásicas/patología , Animales , Neoplasias Encefálicas/metabolismo , Adhesión Celular , Diferenciación Celular , Línea Celular Tumoral , Medios de Cultivo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Trasplante Heterólogo
14.
Biochem Biophys Res Commun ; 360(3): 553-9, 2007 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-17618600

RESUMEN

There is increasing evidence for the presence of cancer stem cells (CSCs) in malignant brain tumors, and these CSCs may play a pivotal role in tumor initiation, growth, and recurrence. Vascular endothelial growth factor (VEGF) promotes the proliferation of vascular endothelial cells (VECs) and the neurogenesis of neural stem cells. Using CSCs derived from human glioblastomas and a retrovirus expressing VEGF, we examined the effects of VEGF on the properties of CSCs in vitro and in vivo. Although VEGF did not affect the property of CSCs in vitro, the injection of mouse brains with VEGF-expressing CSCs led to the massive expansion of vascular-rich GBM, tumor-associated hemorrhage, and high morbidity, suggesting that VEGF promoted tumorigenesis via angiogenesis. These results revealed that VEGF induced the proliferation of VEC in the vascular-rich tumor environment, the so-called stem cell niche.


Asunto(s)
Glioblastoma/irrigación sanguínea , Glioblastoma/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Neovascularización Patológica/inducido químicamente , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Glioblastoma/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...