Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuron ; 109(14): 2275-2291.e8, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34133945

RESUMO

C9orf72 repeat expansions cause inherited amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD) and result in both loss of C9orf72 protein expression and production of potentially toxic RNA and dipeptide repeat proteins. In addition to ALS/FTD, C9orf72 repeat expansions have been reported in a broad array of neurodegenerative syndromes, including Alzheimer's disease. Here we show that C9orf72 deficiency promotes a change in the homeostatic signature in microglia and a transition to an inflammatory state characterized by an enhanced type I IFN signature. Furthermore, C9orf72-depleted microglia trigger age-dependent neuronal defects, in particular enhanced cortical synaptic pruning, leading to altered learning and memory behaviors in mice. Interestingly, C9orf72-deficient microglia promote enhanced synapse loss and neuronal deficits in a mouse model of amyloid accumulation while paradoxically improving plaque clearance. These findings suggest that altered microglial function due to decreased C9orf72 expression directly contributes to neurodegeneration in repeat expansion carriers independent of gain-of-function toxicities.


Assuntos
Envelhecimento/metabolismo , Amiloide/metabolismo , Proteína C9orf72/metabolismo , Microglia/metabolismo , Sinapses/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Amiloide/genética , Animais , Proteína C9orf72/genética , Expansão das Repetições de DNA , Modelos Animais de Doenças , Lisossomos/metabolismo , Camundongos , Camundongos Knockout , Sinapses/patologia
2.
Hum Gene Ther Methods ; 23(4): 271-84, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22950971

RESUMO

Adenoviral vectors (Ads) have been evaluated in clinical trials for glioma. However, systemic immunity against the vectors can hamper therapeutic efficacy. We demonstrated that combined immunostimulation and cytotoxic gene therapy provides long-term survival in preclinical glioma models. Because helper-dependent high-capacity Ads (HC-Ads) elicit sustained transgene expression, in the presence of antiadenoviral immunity, we engineered HC-Ads encoding conditional cytotoxic herpes simplex type 1 thymidine kinase and immunostimulatory cytokine Fms-like tyrosine kinase ligand-3 under the control of the TetOn system. Escalating doses of combined HC-Ads (1×10(8), 1×10(9), and 1×10(10) viral particles [VP]) were delivered into the rat brain. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points after vector delivery. Histopathological analysis did not reveal any evidence of toxicity or long-term inflammation at the lower doses tested. Vector genomes were restricted to the injection site. Serum chemistry did not uncover adverse systemic side effects at any of the doses tested. Taken together, our data indicate that doses of up to 1×10(9) VP of each HC-Ad can be safely administered into the normal brain. This comprehensive toxicity and biodistribution study will lay the foundations for implementation of a phase 1 clinical trial for GBM using HC-Ads.


Assuntos
Adenoviridae/genética , Encéfalo/metabolismo , Vetores Genéticos/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Análise Química do Sangue , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Ensaios Clínicos Fase I como Assunto , Modelos Animais de Doenças , Vetores Genéticos/genética , Vetores Genéticos/toxicidade , Glioma/terapia , Herpesvirus Humano 1/enzimologia , Humanos , Masculino , Ratos , Ratos Endogâmicos Lew , Timidina Quinase/genética , Distribuição Tecidual , Transdução Genética , Tirosina Quinase 3 Semelhante a fms/genética
3.
Neoplasia ; 14(8): 757-70, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22952428

RESUMO

Adenovirus-mediated delivery of the immune-stimulatory cytokine Flt3L and the conditionally cytotoxic thymidine kinase (TK) induces tumor regression and long-term survival in preclinical glioma (glioblastoma multiforme [GBM]) models. Flt3L induces expansion and recruitment of plasmacytoid dendritic cells (pDCs) into the brain. Although pDCs can present antigen and produce powerful inflammatory cytokines, that is, interferon α (IFN-α), their role in tumor immunology remains debated. Thus, we studied the role of pDCs and IFN-α in Ad.TK/GCV+ Ad.Flt3L-mediated anti-GBM therapeutic efficacy. Our data indicate that the combined gene therapy induced recruitment of plasmacytoid DCs (pDCs) into the tumor mass; which were capable of in vivo phagocytosis, IFN-α release, and T-cell priming. Thus, we next used either pDCs or an Ad vector encoding IFN-α delivered within the tumor microenvironment. When rats were treated with Ad.TK/GCV in combination with pDCs or Ad-IFN-α, they exhibited 35% and 50% survival, respectively. However, whereas intracranial administration of Ad.TK/GCV + Ad.Flt3L exhibited a high safety profile, Ad-IFN-α led to severe local inflammation, with neurologic and systemic adverse effects. To elucidate whether the efficacy of the immunotherapy was dependent on IFN-α-secreting pDCs, we administered an Ad vector encoding B18R, an IFN-α antagonist, which abrogated the antitumoral effect of Ad.TK/GCV + Ad.Flt3L. Our data suggest that IFN-α release by activated pDCs plays a critical role in the antitumor effect mediated by Ad.TK/GCV + Ad.Flt3L. In summary, taken together, our results demonstrate that pDCs mediate anti-GBM therapeutic efficacy through the production of IFN-α, thus manipulation of pDCs constitutes an attractive new therapeutic target for the treatment of GBM.


Assuntos
Neoplasias Encefálicas/imunologia , Células Dendríticas/imunologia , Glioblastoma/imunologia , Interferon-alfa/imunologia , Microambiente Tumoral , Adenoviridae/genética , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/patologia , Glioblastoma/terapia , Imunoterapia , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Membrana/genética , Ratos , Linfócitos T/imunologia , Timidina Quinase/genética
4.
Neurotherapeutics ; 9(4): 827-43, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22996231

RESUMO

Immune-mediated gene therapy using adenovirus expressing Flt3 ligand and thymidine kinase followed by ganciclovir administration (Flt3/TK) effectively elicits tumor regression in preclinical glioma models. Herein, we assessed new strategies to optimize Flt3L/TK therapeutic efficacy in a refractory RG2 orthotopic glioblastoma model. Specifically, we aimed to optimize the therapeutic efficacy of Flt3L/TK treatment in the RG2 model by overexpressing the following genes within the brain tumor microenvironment: 1) a TK mutant with enhanced cytotoxicity (SR39 mutant TK), 2) Flt3L-IgG fusion protein that has a longer half-life, 3) CD40L to stimulate DC maturation, 4) T helper cell type 1 polarizing dendritic cell cytokines interleukin-12 or C-X-C motif ligand 10 chemokine (CXCL)-10, 5) C-C motif ligand 2 chemokine (CCL2) or C-C motif ligand 3 chemokine (CCL3) to enhance dendritic cell recruitment into the tumor microenvironment, 6) T helper cell type 1 cytokines interferon-γ or interleukin-2 to enhance effector T-cell functions, and 7) IκBα or p65RHD (nuclear factor kappa-B [NF-κB] inhibitors) to suppress the function of Foxp3+ Tregs and enhanced effector T-cell functions. Anti-tumor immunity and tumor specific effector T-cell functions were assessed by cytotoxic T lymphocyte assay and intracellular IFN-γ staining. Our data showed that overexpression of interferon-γ or interleukin-2, or inhibition of the nuclear factor kappa-B within the tumor microenvironment, enhanced cytotoxic T lymphocyte-mediated immune responses and successfully extended the median survival of rats bearing intracranial RG2 when combined with Flt3L/TK. These findings indicate that enhancement of T-cell functions constitutes a critical therapeutic target to overcome immune evasion and enhance therapeutic efficacy for brain cancer. In addition, our study provides novel targets to be used in combination with immune-therapeutic strategies for glioblastoma, which are currently being tested in the clinic.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Imunoterapia/métodos , Transdução de Sinais , Linfócitos T/imunologia , Adenoviridae/genética , Animais , Antivirais/uso terapêutico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Ganciclovir/uso terapêutico , Vetores Genéticos , Glioblastoma/genética , Glioblastoma/imunologia , Humanos , Interleucina-2/imunologia , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Membrana/uso terapêutico , NF-kappa B/imunologia , Ratos , Proteínas Recombinantes/uso terapêutico , Timidina Quinase/uso terapêutico , Microambiente Tumoral/imunologia
5.
Mol Ther ; 19(10): 1793-801, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21505426

RESUMO

Glioblastoma multiforme (GBM) is a primary brain tumor with a median survival of 14.6 months postdiagnosis. The infiltrative nature of GBM prevents complete resection and residual brain tumor cells give rise to recurrent GBM, a hallmark of this disease. Recurrent GBMs are known to harbor numerous mutations/gene rearrangements when compared to the primary tumor, which leads to the potential expression of novel proteins that could serve as tumor neoantigens. We have developed a combined immune-based gene therapeutic approach for GBM using adenoviral (Ads) mediated gene delivery of Herpes Simplex Virus Type 1-thymidine kinase (TK) into the tumor mass to induce tumor cells' death combined with an adenovirus expressing fms-like tyrosine kinase 3 ligand (Flt3L) to recruit dendritic cells (DCs) into the tumor microenvironment. This leads to the induction of specific anti-brain tumor immunity and immunological memory. In a model of GBM recurrence, we demonstrate that Flt3L/TK mediated immunological memory is capable of recognizing brain tumor neoantigens absent from the original treated tumor. These data demonstrate that the Flt3L/TK gene therapeutic approach can induce systemic immunological memory capable of recognizing a brain tumor neoantigen in a model of recurrent GBM.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/terapia , Timidina Quinase/genética , Tirosina Quinase 3 Semelhante a fms/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Memória Imunológica , Interferon gama/metabolismo , Linfócitos T/imunologia
6.
Discov Med ; 10(53): 293-304, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21034670

RESUMO

Glioblastoma multiforme (GBM) is a deadly primary brain tumor in adults, with a median survival of ~12-18 months post-diagnosis. Despite recent advances in conventional therapeutic approaches, only modest improvements in median survival have been achieved; GBM usually recurs within 12 months post-resection, with poor prognosis. Thus, novel therapeutic strategies to target and kill GBM cells are desperately needed. Our group and others are pursuing virotherapy and gene therapy strategies for the treatment of GBM. In this review, we will discuss various virotherapy and gene therapy approaches for GBM currently under pre-clinical and clinical evaluation including direct or conditional cytotoxic, and/or immunostimulatory approaches. We also discuss cutting-edge technologies for drug/gene delivery and targeting brain tumors, including the use of stem cells as delivery platforms, the use of targeted immunotoxins, and the therapeutic potential of using GBM microvesicles to deliver therapeutic siRNAs or virotherapies. Finally, various animal models available to test novel GBM therapies are discussed.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Terapia Viral Oncolítica/métodos , Terapias em Estudo/métodos , Adulto , Animais , Terapia Genética/tendências , Humanos , Modelos Biológicos , Terapia Viral Oncolítica/tendências , Terapias em Estudo/tendências
7.
PLoS One ; 5(6): e11074, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20552015

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and carries a dismal prognosis. We have developed a conditional cytotoxic/immunotherapeutic approach using adenoviral vectors (Ads) encoding the immunostimulatory cytokine, human soluble fms-like tyrosine kinase 3 ligand (hsFlt3L) and the conditional cytotoxic molecule, i.e., Herpes Simplex Type 1- thymide kinase (TK). This therapy triggers an anti-tumor immune response that leads to tumor regression and anti-tumor immunological memory in intracranial rodent cancer models. We aim to test the efficacy of this immunotherapy in dogs bearing spontaneous GBM. In view of the controversy regarding the effect of human cytokines on dog immune cells, and considering that the efficacy of this treatment depends on hsFlt3L-stimulated dendritic cells (DCs), in the present work we tested the ability of Ad-encoded hsFlt3L to generate DCs from dog peripheral blood and compared its effects with canine IL-4 and GM-CSF. METHODOLOGY/PRINCIPAL FINDINGS: Our results demonstrate that hsFlT3L expressed form an Ad vector, generated DCs from peripheral blood cultures with very similar morphological and phenotypic characteristics to canine IL-4 and GM-CSF-cultured DCs. These include phagocytic activity and expression of CD11c, MHCII, CD80 and CD14. Maturation of DCs cultured under both conditions resulted in increased secretion of IL-6, TNF-alpha and IFN-gamma. Importantly, hsFlt3L-derived antigen presenting cells showed allostimulatory potential highlighting their ability to present antigen to T cells and elicit their proliferation. CONCLUSIONS/SIGNIFICANCE: These results demonstrate that hsFlt3L induces the proliferation of canine DCs and support its use in upcoming clinical trials for canine GBM. Our data further support the translation of hsFlt3L to be used for dendritic cells' vaccination and gene therapeutic approaches from rodent models to canine patients and its future implementation in human clinical trials.


Assuntos
Neoplasias Encefálicas/veterinária , Células Dendríticas/imunologia , Doenças do Cão/terapia , Glioma/veterinária , Imunoterapia , Proteínas de Membrana/imunologia , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Proliferação de Células , Células Cultivadas , Doenças do Cão/imunologia , Cães , Vetores Genéticos , Glioma/imunologia , Glioma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Herpesvirus Humano 1/genética , Interleucina-4/administração & dosagem , Ativação Linfocitária , Proteínas de Membrana/genética , Fagocitose
8.
Clin Cancer Res ; 15(13): 4401-14, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19570774

RESUMO

PURPOSE: In preparation for a phase I clinical trial using a combined cytotoxic/immunotherapeutic strategy with adenoviruses (Ad) expressing Flt3L (Ad-Flt3L) and thymidine kinase (Ad-TK) to treat glioblastoma (GBM), we tested the hypothesis that Ad-TK+GCV would be the optimal tumor-killing agent in relation to efficacy and safety when compared with other proapoptotic approaches. EXPERIMENTAL DESIGN: The efficacy and neurotoxicity of Ad-TK+GCV was compared with Ads encoding the proapoptotic cytokines [tumor necrosis factor-alpha, tumor necrosis factor-related apoptosis-inducing factor (TRAIL), and Fas ligand (FasL)], alone or in combination with Ad-Flt3L. In rats bearing small GBMs (day 4), only Ad-TK+GCV or Ad-FasL improved survival. RESULTS: In rats bearing large GBMs (day 9), the combination of Ad-Flt3L with Ad-FasL did not improve survival over FasL alone, whereas Ad-Flt3L combined with Ad-TK+GCV led to 70% long-term survival. Expression of FasL and TRAIL caused severe neuropathology, which was not encountered when we used Ad-TK+/-Ad-Flt3L. In vitro, all treatments elicited release of high mobility group box 1 protein (HMGB1) from dying tumor cells. In vivo, the highest levels of circulating HMGB1 were observed after treatment with Ad-TK+GCV+Ad-Flt3L; HMGB1 was necessary for the therapeutic efficacy of AdTK+GCV+Ad-Flt3L because its blockade with glycyrrhizin completely blocked tumor regression. We also showed the killing efficacy of Ad-TK+GCV in human GBM cell lines and GBM primary cultures, which also elicited release of HMGB1. CONCLUSIONS: Our results indicate that Ad-TK+GCV+Ad-Flt3L exhibit the highest efficacy and safety profile among the several proapoptotic approaches tested. The results reported further support the implementation of this combined approach in a phase I clinical trial for GBM.


Assuntos
Apoptose/fisiologia , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/terapia , Glioma/sangue , Glioma/terapia , Proteína HMGB1/sangue , Neurônios/patologia , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Terapia Combinada , Glioma/metabolismo , Glioma/patologia , Proteína HMGB1/metabolismo , Transplante de Neoplasias , Neurônios/virologia , Terapia Viral Oncolítica/efeitos adversos , Ratos , Ratos Endogâmicos Lew , Timidina Quinase/genética , Transplante Isogênico , Resultado do Tratamento , Células Tumorais Cultivadas , Tirosina Quinase 3 Semelhante a fms/genética
9.
PLoS Med ; 6(1): e10, 2009 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-19143470

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor that carries a 5-y survival rate of 5%. Attempts at eliciting a clinically relevant anti-GBM immune response in brain tumor patients have met with limited success, which is due to brain immune privilege, tumor immune evasion, and a paucity of dendritic cells (DCs) within the central nervous system. Herein we uncovered a novel pathway for the activation of an effective anti-GBM immune response mediated by high-mobility-group box 1 (HMGB1), an alarmin protein released from dying tumor cells, which acts as an endogenous ligand for Toll-like receptor 2 (TLR2) signaling on bone marrow-derived GBM-infiltrating DCs. METHODS AND FINDINGS: Using a combined immunotherapy/conditional cytotoxic approach that utilizes adenoviral vectors (Ad) expressing Fms-like tyrosine kinase 3 ligand (Flt3L) and thymidine kinase (TK) delivered into the tumor mass, we demonstrated that CD4(+) and CD8(+) T cells were required for tumor regression and immunological memory. Increased numbers of bone marrow-derived, tumor-infiltrating myeloid DCs (mDCs) were observed in response to the therapy. Infiltration of mDCs into the GBM, clonal expansion of antitumor T cells, and induction of an effective anti-GBM immune response were TLR2 dependent. We then proceeded to identify the endogenous ligand responsible for TLR2 signaling on tumor-infiltrating mDCs. We demonstrated that HMGB1 was released from dying tumor cells, in response to Ad-TK (+ gancyclovir [GCV]) treatment. Increased levels of HMGB1 were also detected in the serum of tumor-bearing Ad-Flt3L/Ad-TK (+GCV)-treated mice. Specific activation of TLR2 signaling was induced by supernatants from Ad-TK (+GCV)-treated GBM cells; this activation was blocked by glycyrrhizin (a specific HMGB1 inhibitor) or with antibodies to HMGB1. HMGB1 was also released from melanoma, small cell lung carcinoma, and glioma cells treated with radiation or temozolomide. Administration of either glycyrrhizin or anti-HMGB1 immunoglobulins to tumor-bearing Ad-Flt3L and Ad-TK treated mice, abolished therapeutic efficacy, highlighting the critical role played by HMGB1-mediated TLR2 signaling to elicit tumor regression. Therapeutic efficacy of Ad-Flt3L and Ad-TK (+GCV) treatment was demonstrated in a second glioma model and in an intracranial melanoma model with concomitant increases in the levels of circulating HMGB1. CONCLUSIONS: Our data provide evidence for the molecular and cellular mechanisms that support the rationale for the clinical implementation of antibrain cancer immunotherapies in combination with tumor killing approaches in order to elicit effective antitumor immune responses, and thus, will impact clinical neuro-oncology practice.


Assuntos
Neoplasias Encefálicas/metabolismo , Proteína HMGB1/metabolismo , Receptor 2 Toll-Like/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Citometria de Fluxo , Vetores Genéticos , Glioblastoma/imunologia , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Transgênicos
10.
Mol Ther ; 16(4): 682-90, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18283279

RESUMO

Glioblastoma multiforme (GBM) is an invasive and aggressive primary brain tumor which is associated with a dismal prognosis. We have earlier developed a macroscopic, intracranial, syngeneic GBM model, in which treatment with adenoviral vectors (Ads) expressing herpes simplex virus type 1 thymidine kinase (HSV1-TK) plus ganciclovir (GCV) resulted in survival of approximately 20% of the animals. In this model, treatment with Ads expressing Fms-like tyrosine kinase 3 ligand (Flt3L), in combination with Ad-HSV1-TK improves the survival rate to approximately 70% and induces systemic antitumor immunity. We hypothesized that the growth of a large intracranial tumor mass would cause behavioral abnormalities that can be reversed by the combined gene therapy. We assessed the behavior and neuropathology of tumor-bearing animals treated with the combined gene therapy, 3 days after treatment, in long-term survivors, and in a recurrent model of glioma. We demonstrate that the intracranial GBM induces behavioral deficits that are resolved after treatment with Ad-Flt3L/Ad-TK (+GCV). Neuropathological analysis of long-term survivors revealed an overall recovery of normal brain architecture. The lack of long-term behavioral deficits and limited neuropathological abnormalities demonstrate the efficacy and safety of the combined Ad-Flt3L/Ad-TK gene therapy for GBM. These findings can serve to underpin further developments of this therapeutic modality, leading toward implementation of a Phase I clinical trial.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Herpesvirus Humano 1/enzimologia , Proteínas de Membrana/genética , Timidina Quinase/genética , Adenoviridae/metabolismo , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Terapia Genética , Vetores Genéticos , Glioblastoma/patologia , Glioblastoma/fisiopatologia , Masculino , Proteínas de Membrana/biossíntese , Atividade Motora , Transplante de Neoplasias , Ratos , Ratos Endogâmicos Lew , Comportamento Estereotipado , Timidina Quinase/metabolismo
11.
J Virol ; 82(9): 4680-4, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18287240

RESUMO

Gene therapy is proposed as a novel therapeutic strategy for treating glioblastoma multiforme (GBM), a devastating brain cancer. In the clinic, antivector immune responses pose formidable challenges. Herein we demonstrate that high-capacity adenovirus vectors (HC-Ads) carrying the conditional cytotoxic gene herpes simplex virus type 1-thymidine kinase (TK) induce tumor regression and long-term survival in an intracranial glioma model, even in the presence of systemic antiadenovirus immunity, as could be encountered in patients. First-generation Ad-TK failed to elicit tumor regression in this model. These results pave the way for implementing HC-Ad-TK-mediated gene therapy as a powerful adjuvant for treating GBM.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Glioblastoma/terapia , Timidina Quinase/uso terapêutico , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/farmacologia , Encéfalo/patologia , Neoplasias Encefálicas/terapia , Modelos Animais de Doenças , Herpesvirus Humano 1/enzimologia , Humanos , Ratos , Ratos Endogâmicos Lew , Taxa de Sobrevida , Timidina Quinase/genética , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos
12.
Neuro Oncol ; 10(1): 19-31, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18079358

RESUMO

The disseminated characteristics of human glioblastoma multiforme (GBM) make it a particularly difficult tumor to treat with long-term efficacy. Most preclinical models of GBM involve treatment of a single tumor mass. For therapeutic outcomes to translate from the preclinical to the clinical setting, induction of an antitumor response capable of eliminating multifocal disease is essential. We tested the hypothesis that expression of Flt3L (human soluble FMS-like tyrosine kinase 3 ligand) and TK (herpes simplex virus type 1-thymidine kinase) within brain gliomas would mediate regression of the primary, treated tumor mass and a secondary, untreated tumor growing at a distant site from the primary tumor and the site of therapeutic vector injection. In both the single-GBM and multifocal-GBM models used, all saline-treated control animals succumbed to tumors by day 22. Around 70% of the animals bearing a single GBM mass treated with an adenovirus expressing Flt3L (AdFlt3L) and an adenovirus expressing TK (AdTK + GCV) survived long term. Approximately 50% of animals bearing a large primary GBM that were implanted with a second GBM in the contralateral hemisphere at the same time the primary tumors were being treated with AdFlt3L and AdTK also survived long term. A second multifocal GBM model, in which bilateral GBMs were implanted simultaneously and only the right tumor mass was treated with AdFlt3L and AdTK, also demonstrated long-term survival. While no significant difference in survival was found between unifocal and multifocal GBM-bearing animals treated with AdFlt3L and AdTK, both treatments were statistically different from the saline-treated control group (p < 0.05). Our results demonstrate that combination therapy with AdFlt3L and AdTK can eradicate multifocal brain tumor disease in a syngeneic, intracranial GBM model.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Proteínas de Membrana/metabolismo , Timidina Quinase/metabolismo , Adenoviridae/genética , Animais , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Glioblastoma/patologia , Humanos , Masculino , Microscopia Confocal , Ratos , Ratos Endogâmicos Lew , Transplante Isogênico
13.
Neuro Oncol ; 9(3): 245-58, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17522335

RESUMO

Expression of the immune-stimulatory molecule Fms-like tyrosine kinase 3 ligand (Flt3L) and the conditional cytotoxic enzyme herpes simplex virus type 1 thymidine kinase (HSV1-TK) provides long-term immune-mediated survival of large glioblastoma multiforme (GBM) models in rodents. A limitation for predictive testing of novel antiglioma therapies has been the lack of a glioma model in a large animal. Dogs bearing spontaneous GBM may constitute an attractive large-animal model for GBM, which so far has remained underappreciated. In preparation for a clinical trial in dogs bearing spontaneous GBMs, we tested and optimized adenovirus-mediated transgene expression with negligible toxicity in the dog brain in vivo and in canine J3T glioma cells. Expression of the marker gene beta-galactosidase (beta-Gal) was higher when driven by the murine (m) than the human (h) cytomegalovirus (CMV) promoter in the dog brain in vivo, without enhanced inflammation. In the canine brain, beta-Gal was expressed mostly in astrocytes. beta-Gal activity in J3T cells was also higher with the mCMV than the hCMV promoter driving tetracycline-dependent (TetON) transgene expression within high-capacity adenovirus vectors (HC-Ads). Dog glioma cells were efficiently transduced by HC-Ads expressing mCMV-driven HSV1-TK, which induced 90% reduction in cell viability in the presence of ganciclovir. J3T cells were also effectively transduced with HC-Ads expressing Flt3L under the control of the regulatable TetON promoter system, and as predicted, Flt3L release was stringently inducer dependent. HC-Ads encoding therapeutic transgenes under the control of regulatory sequences driven by the mCMV promoter are excellent vectors for the treatment of spontaneous GBM in dogs, which constitute an ideal preclinical animal model.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/genética , Encéfalo/fisiologia , Terapia Genética/métodos , Glioma/genética , Regiões Promotoras Genéticas , Transgenes/fisiologia , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Citomegalovirus/genética , Cães , Ensaio de Imunoadsorção Enzimática , Engenharia Genética/métodos , Vetores Genéticos , Glioma/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Microscopia Confocal , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Transdução Genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA