Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(20): 7835-40, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22547816

RESUMO

Following antigen recognition on target cells, effector T cells establish immunological synapses and secrete cytokines. It is thought that T cells secrete cytokines in one of two modes: either synaptically (i.e., toward antigenic target cells) or multidirectionally, affecting a wider population of cells. This paradigm predicts that synaptically secreted cytokines such as IFN-γ will preferentially signal to antigenic target cells contacted by the T cell through an immunological synapse. Despite its physiological significance, this prediction has never been tested. We developed a live-cell imaging system to compare the responses of target cells and nonantigenic bystanders to IFN-γ secreted by CD8+, antigen-specific, cytotoxic T cells. Both target cells and surrounding nontarget cells respond robustly. This pattern of response was detected even at minimal antigenic T-cell stimulation using low doses of antigenic peptide, or altered peptide ligands. Although cytotoxic immunological synapses restrict killing to antigenic target cells, the effects of IFN-γ are more widespread.


Assuntos
Sinapses Imunológicas/imunologia , Interferon gama/metabolismo , Linfócitos T Citotóxicos/imunologia , Adenoviridae , Análise de Variância , Astrócitos/imunologia , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Processamento de Imagem Assistida por Computador , Interferon gama/imunologia , Microscopia/métodos
2.
Toxicol Appl Pharmacol ; 268(3): 318-30, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23403069

RESUMO

Adenoviral vectors (Ads) are promising gene delivery vehicles due to their high transduction efficiency; however, their clinical usefulness has been hampered by their immunogenicity and the presence of anti-Ad immunity in humans. We reported the efficacy of a gene therapy approach for glioma consisting of intratumoral injection of Ads encoding conditionally cytotoxic herpes simplex type 1 thymidine kinase (Ad-TK) and the immunostimulatory cytokine fms-like tyrosine kinase ligand 3 (Ad-Flt3L). Herein, we report the biodistribution, efficacy, and neurological and systemic effects of a bicistronic high-capacity Ad, i.e., HC-Ad-TK/TetOn-Flt3L. HC-Ads elicit sustained transgene expression, even in the presence of anti-Ad immunity, and can encode large therapeutic cassettes, including regulatory elements to enable turning gene expression "on" or "off" according to clinical need. The inclusion of two therapeutic transgenes within a single vector enables a reduction of the total vector load without adversely impacting efficacy. Because clinically the vectors will be delivered into the surgical cavity, normal regions of the brain parenchyma are likely to be transduced. Thus, we assessed any potential toxicities elicited by escalating doses of HC-Ad-TK/TetOn-Flt3L (1×10(8), 1×10(9), or 1×10(10) viral particles [vp]) delivered into the rat brain parenchyma. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points. The results indicate that doses up to 1×10(9) vp of HC-Ad-TK/TetOn-Flt3L can be safely delivered into the normal rat brain and underpin further developments for its implementation in a phase I clinical trial for glioma.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Ensaios Clínicos Fase I como Assunto/métodos , Citotoxinas/administração & dosagem , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Glioblastoma/tratamento farmacológico , Imunização/métodos , Adenoviridae/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Citotoxinas/efeitos adversos , Citotoxinas/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Quimioterapia Combinada , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Vetores Genéticos/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Ratos , Ratos Endogâmicos Lew , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia , Resultado do Tratamento
3.
Mol Ther ; 20(4): 808-19, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22233583

RESUMO

The adaptive immune response to viral vectors reduces vector-mediated transgene expression from the brain. It is unknown, however, whether this loss is caused by functional downregulation of transgene expression or death of transduced cells. Herein, we demonstrate that during the elimination of transgene expression, the brain becomes infiltrated with CD4(+) and CD8(+) T cells and that these T cells are necessary for transgene elimination. Further, the loss of transgene-expressing brain cells fails to occur in the absence of IFNγ, perforin, and TNFα receptor. Two methods to induce severe immune suppression in immunized animals also fail to restitute transgene expression, demonstrating the irreversibility of this process. The need for cytotoxic molecules and the irreversibility of the reduction in transgene expression suggested to us that elimination of transduced cells is responsible for the loss of transgene expression. A new experimental paradigm that discriminates between downregulation of transgene expression and the elimination of transduced cells demonstrates that transduced cells are lost from the brain upon the induction of a specific antiviral immune response. We conclude that the anti-adenoviral immune response reduces transgene expression in the brain through loss of transduced cells.


Assuntos
Encéfalo/citologia , Interferon gama/metabolismo , Perforina/metabolismo , Transdução Genética/métodos , Transgenes/genética , Fator de Necrose Tumoral alfa/metabolismo , Adenoviridae/genética , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Transgênicos
4.
Proc Natl Acad Sci U S A ; 107(46): 20021-6, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21030678

RESUMO

Restricting the cytotoxicity of anticancer agents by targeting receptors exclusively expressed on tumor cells is critical when treating infiltrative brain tumors such as glioblastoma multiforme (GBM). GBMs express an IL-13 receptor (IL13Rα2) that differs from the physiological IL4R/IL13R receptor. We developed a regulatable adenoviral vector (Ad.mhIL-4.TRE.mhIL-13-PE) encoding a mutated human IL-13 fused to Pseudomonas exotoxin (mhIL-13-PE) that specifically binds to IL13Rα2 to provide sustained expression, effective anti-GBM cytotoxicity, and minimal neurotoxicity. The therapeutic Ad also encodes mutated human IL-4 that binds to the physiological IL4R/IL13R without interacting with IL13Rα2, thus inhibiting potential binding of mhIL-13-PE to normal brain cells. Using intracranial GBM xenografts and syngeneic mouse models, we tested the Ad.mhIL-4.TRE.mhIL-13-PE and two protein formulations, hIL-13-PE used in clinical trials (Cintredekin Besudotox) and a second-generation mhIL-13-PE. Cintredekin Besudotox doubled median survival without eliciting long-term survival and caused severe neurotoxicity; mhIL-13-PE led to ∼40% long-term survival, eliciting severe neurological toxicity at the high dose tested. In contrast, Ad-mediated delivery of mhIL-13-PE led to tumor regression and long-term survival in over 70% of the animals, without causing apparent neurotoxicity. Although Cintredekin Besudotox was originally developed to target GBM, when tested in a phase III trial it failed to achieve clinical endpoints and revealed neurotoxicity. Limitations of Cintredekin Besudotox include its short half-life, which demanded frequent or continued administration, and binding to IL4R/IL13R, present in normal brain cells. These shortcomings were overcome by our therapeutic Ad, thus representing a significant advance in the development of targeted therapeutics for GBM.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Citotoxinas/genética , Citotoxinas/uso terapêutico , Técnicas de Transferência de Genes , Terapia Genética , Glioma/tratamento farmacológico , Adenoviridae/genética , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Exotoxinas/genética , Exotoxinas/uso terapêutico , Vetores Genéticos/genética , Glioma/patologia , Humanos , Imunocompetência/imunologia , Interleucina-13/genética , Interleucina-13/uso terapêutico , Camundongos , Camundongos Nus , Mutação/genética , Neurotoxinas/toxicidade , Pseudomonas/metabolismo , Transgenes/genética , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Proc Natl Acad Sci U S A ; 107(32): 14443-8, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20660723

RESUMO

Soluble antigens diffuse out of the brain and can thus stimulate a systemic immune response, whereas particulate antigens (from infectious agents or tumor cells) remain within brain tissue, thus failing to stimulate a systemic immune response. Immune privilege describes how the immune system responds to particulate antigens localized selectively within the brain parenchyma. We believe this immune privilege is caused by the absence of antigen presenting dendritic cells from the brain. We tested the prediction that expression of fms-like tyrosine kinase ligand 3 (Flt3L) in the brain will recruit dendritic cells and induce a systemic immune response against exogenous influenza hemagglutinin in BALB/c mice. Coexpression of Flt3L with HA in the brain parenchyma induced a robust systemic anti-HA immune response, and a small response against myelin basic protein and proteolipid protein epitopes. Depletion of CD4(+)CD25+ regulatory T cells (Tregs) enhanced both responses. To investigate the autoimmune impact of these immune responses, we characterized the neuropathological and behavioral consequences of intraparenchymal injections of Flt3L and HA in BALB/c and C57BL/6 mice. T cell infiltration in the forebrain was time and strain dependent, and increased in animals treated with Flt3L and depleted of Tregs; however, we failed to detect widespread defects in myelination throughout the forebrain or spinal cord. Results of behavioral tests were all normal. These results demonstrate that Flt3L overcomes the brain's immune privilege, and supports the clinical development of Flt3L as an adjuvant to stimulate clinically effective immune responses against brain neo-antigens, for example, those associated with brain tumors.


Assuntos
Encéfalo/imunologia , Sistema Imunitário/imunologia , Tirosina Quinase 3 Semelhante a fms/imunologia , Adjuvantes Imunológicos , Animais , Antígenos/imunologia , Células Dendríticas/imunologia , Hemaglutininas/imunologia , Imunidade , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Prosencéfalo/imunologia , Medula Espinal/imunologia , Linfócitos T Reguladores/imunologia
6.
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
7.
J Virol ; 84(12): 6007-17, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375153

RESUMO

Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Conditional cytotoxic/immune-stimulatory gene therapy (Ad-TK and Ad-Flt3L) elicits tumor regression and immunological memory in rodent GBM models. Since the majority of patients enrolled in clinical trials would exhibit adenovirus immunity, which could curtail transgene expression and therapeutic efficacy, we used high-capacity adenovirus vectors (HC-Ads) as a gene delivery platform. Herein, we describe for the first time a novel bicistronic HC-Ad driving constitutive expression of herpes simplex virus type 1 thymidine kinase (HSV1-TK) and inducible Tet-mediated expression of Flt3L within a single-vector platform. We achieved anti-GBM therapeutic efficacy with no overt toxicities using this bicistronic HC-Ad even in the presence of systemic Ad immunity. The bicistronic HC-Ad-TK/TetOn-Flt3L was delivered into intracranial gliomas in rats. Survival, vector biodistribution, neuropathology, systemic toxicity, and neurobehavioral deficits were assessed for up to 1 year posttreatment. Therapeutic efficacy was also assessed in animals preimmunized against Ads. We demonstrate therapeutic efficacy, with vector genomes being restricted to the brain injection site and an absence of overt toxicities. Importantly, antiadenoviral immunity did not inhibit therapeutic efficacy. These data represent the first report of a bicistronic vector platform driving the expression of two therapeutic transgenes, i.e., constitutive HSV1-TK and inducible Flt3L genes. Further, our data demonstrate no promoter interference and optimum gene delivery and expression from within this single-vector platform. Analysis of the efficacy, safety, and toxicity of this bicistronic HC-Ad vector in an animal model of GBM strongly supports further preclinical testing and downstream process development of HC-Ad-TK/TetOn-Flt3L for a future phase I clinical trial for GBM.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Glioma/genética , Glioma/terapia , Herpesvirus Humano 1/enzimologia , Timidina Quinase/uso terapêutico , Proteínas Virais/uso terapêutico , Tirosina Quinase 3 Semelhante a fms/uso terapêutico , Adenoviridae/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação da Expressão Gênica , Terapia Genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glioma/metabolismo , Herpesvirus Humano 1/genética , Humanos , Ratos , Ratos Endogâmicos Lew , Timidina Quinase/genética , Timidina Quinase/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
8.
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
9.
J Virol ; 82(20): 9978-93, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684835

RESUMO

The clearance of virally infected cells from the brain is mediated by T cells that engage antigen-presenting cells to form supramolecular activation clusters at the immunological synapse. However, after clearance, the T cells persist at the infection site and remain activated locally. In the present work the long-term interactions of immune cells in brains of monkeys were imaged in situ 9 months after the viral inoculation. After viral immunity, the persistent infiltration of T cells and B cells was observed at the infection sites. T cells showed evidence of T-cell receptor signaling as a result of contacts with B cells. Three-dimensional analysis of B-cell-T-cell synapses showed clusters of CD3 in T cells and the segregation of CD20 in B cells, involving the recruitment of CD40 ligand at the interface. These results demonstrate that immunological synapses between B cells and T cells forming three-dimensional microclusters occur in vivo in the central nervous system and suggest that these interactions may be involved in the lymphocyte activation after viral immunity at the original infection site.


Assuntos
Antígenos CD20/imunologia , Linfócitos B/imunologia , Encéfalo/imunologia , Complexo CD3/imunologia , Ligante de CD40/imunologia , Primatas/imunologia , Linfócitos T/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Infecções por Adenoviridae/imunologia , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Linfócitos B/citologia , Encéfalo/anatomia & histologia , Comunicação Celular/fisiologia , Feminino , Ativação Linfocitária/imunologia , Cooperação Linfocítica/imunologia , Macaca fascicularis , Masculino , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/citologia
10.
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
11.
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
12.
Mol Ther ; 16(2): 343-51, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18180781

RESUMO

Immune responses against vectors or encoded transgenes can impose limitations on gene therapy. We demonstrated that tetracycline-regulated high-capacity adenoviral vectors (HC-Ads) sustain regulated transgene expression in the brain even in the presence of systemic pre-existing immune responses against adenoviruses. In this study we assessed whether systemic pre-existing immune responses against the transgene products, i.e., beta-Gal or the tetracycline-dependent (TetON) regulatory transcription factors (rtTA2(S)M2 and the tTS(Kid)), affect transgene expression levels and the safety profile of HC-Ads in the brain. We pre-immunized mice with plasmids encoding the TetON switch expressing rtTA2(S)M2 and the tTS(Kid) or beta-Gal. HC-Ads expressing beta-Gal under the control of the TetON switch were then injected into the striatum. We assessed levels and distribution of beta-Gal expression, and evaluated local inflammation and neuropathological changes. We found that systemic immunity against beta-Gal, but not against the TetON switch, led to inflammation and reduction of transgene expression in the striatum. Therefore, the regulatory TetON switch appears to be safe to use, and capable of sustaining transgene expression in the brain even in the presence of an immune response against its components. Systemic immunity against the transgene had the effect of curtailing its expression, thereby affecting the efficacy and safety of gene delivery to the brain. This factor should be considered when developing gene therapies for neurological use.


Assuntos
Adenoviridae/genética , Encéfalo/metabolismo , Imunização/métodos , Transgenes/genética , Animais , Western Blotting , Encéfalo/imunologia , Feminino , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Imuno-Histoquímica , Inflamação/imunologia , Camundongos , Plasmídeos/genética , Tetraciclina/farmacologia , beta-Galactosidase/metabolismo
13.
Mol Ther ; 16(4): 682-690, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28178463

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 ∼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 ∼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.

14.
J Neurosurg ; 107(3): 568-77, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17886557

RESUMO

OBJECT: A hollow fiber catheter was developed to improve the distribution of drugs administered via direct infusion into the central nervous system (CNS). It is a porous catheter that significantly increases the surface area of brain tissue into which a drug is infused. METHODS: Dye was infused into the mouse brain through convection-enhanced delivery (CED) using a 28-gauge needle compared with a 3-mm-long hollow fiber catheter. To determine whether a hollow fiber catheter could increase the distribution of gene therapy vectors, a recombinant adenovirus expressing the firefly luciferase reporter was injected into the mouse striatum. Gene expression was monitored using in vivo bioluminescent imaging. To assess the distribution of gene transfer, an adenovirus expressing green fluorescent protein was injected into the striatum using a hollow fiber catheter or a needle. RESULTS: Hollow fiber catheter-mediated infusion increased the volume of brain tissue labeled with dye by 2.7 times relative to needle-mediated infusion. In vivo imaging revealed that catheter-mediated infusion of adenovirus resulted in gene expression that was 10-times greater than that mediated by a needle. The catheter appreciably increased the area of brain transduced with adenovirus relative to a needle, affecting a significant portion of the injected hemisphere. CONCLUSIONS: The miniature hollow fiber catheter used in this study significantly increased the distribution of dye and adenoviral-mediated gene transfer in the mouse brain compared with the levels reached using a 28-gauge needle. Compared with standard single-port clinical catheters, the hollow fiber catheter has the advantage of millions of nanoscale pores to increase surface area and bulk flow in the CNS. Extending the scale of the hollow fiber catheter for the large mammalian brain shows promise in increasing the distribution and efficacy of gene therapy and drug therapy using CED.


Assuntos
Adenoviridae , Encéfalo/metabolismo , Cateterismo , Técnicas de Transferência de Genes/instrumentação , Vetores Genéticos/farmacocinética , Adenoviridae/enzimologia , Adenoviridae/genética , Animais , Materiais Biocompatíveis , Corantes/administração & dosagem , Corantes/farmacocinética , Convecção , Desenho de Equipamento , Azul Evans/administração & dosagem , Azul Evans/farmacocinética , Vetores Genéticos/administração & dosagem , Infusões Parenterais/instrumentação , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos CBA , Polímeros , Sulfonas
15.
J Neurosurg ; 107(1): 136-44, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17639883

RESUMO

OBJECT: The purpose of this study was to evaluate the gene transfer capability and tolerability of plasmid DNA/polyethylenimine (PEI) complexes in comparison with adenovirus and naked plasmid DNA in the canine brain. METHODS: Plasmid or adenoviral vectors encoding firefly luciferase were injected directly into the cerebral parenchyma of five adult dogs at varying doses and volumes. Serial physical and neurological examinations, as well as blood and cerebrospinal fluid (CSF) analyses, were conducted before and after the surgery for 3 days. Three days after gene delivery, a luciferase activity assay and immunofluorescence analysis were used to test the brain tissue for gene expression. RESULTS: Injection into the brain parenchyma resulted in gene transfer throughout the cerebrum with every vector tested. Luciferase expression was highest when adenovirus vectors were used. Injection of plasmid DNA/PEI complexes and naked DNA resulted in similar levels of luciferase expression, which were on average 0.5 to 1.5% of the expression achieved with adenovirus vectors. Immunofluorescent microscopy analysis revealed that plasmid DNA/PEI complexes transduced mainly neurons, whereas adenovirus transduced mainly astrocytes. No significant acute side effects or neurological complications were observed in any of the dogs. Mononuclear cell counts significantly increased in the CSF after adenovirus injection and modestly increased after injection of plasmid DNA/PEI complexes, suggesting that a mild, acute inflammatory response occurred in the central nervous system (CNS). CONCLUSIONS: Compared with rodent models that are limited by very small brains, the dog is an excellent preclinical model in which to assess the distribution and safety of emerging gene transfer technologies. In this study, short-term gene transfer was evaluated as a prelude to long-term expression and safety studies. The authors conclude that the viral and nonviral vectors tested were well tolerated and effective at mediating gene transfer throughout a large portion of the canine brain. The nonviral plasmid vectors were less effective than adenovirus, yet they still achieved appreciable gene expression levels. Due to reduced gene transfer efficiency relative to viral vectors, nonviral vectors may be most useful when the expressed protein is secreted or exerts a bystander effect. Nonviral vectors offer an alternative means to genetically modify cells within the CNS of large mammals.


Assuntos
Adenovirus Caninos/genética , Técnicas de Transferência de Genes/instrumentação , Terapia Genética/instrumentação , Plasmídeos/genética , Animais , Astrócitos/citologia , Astrócitos/virologia , Análise Química do Sangue , Encéfalo/citologia , Encéfalo/enzimologia , Encéfalo/virologia , Neoplasias Encefálicas/terapia , Viroses do Sistema Nervoso Central/genética , Viroses do Sistema Nervoso Central/patologia , Viroses do Sistema Nervoso Central/virologia , Cães , Estudos de Viabilidade , Vetores Genéticos/genética , Glioma/terapia , Inflamação/patologia , Inflamação/virologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Microscopia de Fluorescência , Neurônios/citologia , Neurônios/metabolismo , Neurônios/virologia , Plasmídeos/fisiologia , Polietilenoimina/uso terapêutico , Transdução Genética/métodos , Vacinas de DNA/genética
16.
Curr Top Med Chem ; 5(12): 1151-70, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16248789

RESUMO

Glioblastoma (GBM) is a type of intracranial brain tumor, for which there is no cure. In spite of advances in surgery, chemotherapy and radiotherapy, patients die within a year of diagnosis. Therefore, there is a critical need to develop novel therapeutic approaches for this disease. Gene therapy, which is the use of genes or other nucleic acids as drugs, is a powerful new treatment strategy which can be developed to treat GBM. Several treatment modalities are amenable for gene therapy implementation, e.g. conditional cytotoxic approaches, targeted delivery of toxins into the tumor mass, immune stimulatory strategies, and these will all be the focus of this review. Both conditional cytotoxicity and targeted toxin mediated tumor death, are aimed at eliminating an established tumor mass and preventing further growth. Tumors employ several defensive strategies that suppress and inhibit anti-tumor immune responses. A better understanding of the mechanisms involved in eliciting anti-tumor immune responses has identified promising targets for immunotherapy. Immunotherapy is designed to aid the immune system to recognize and destroy tumor cells in order to eliminate the tumor burden. Also, immune-therapeutic strategies have the added advantage that an activated immune system has the capability of recognizing tumor cells at distant sites from the primary tumor, therefore targeting metastasis distant from the primary tumor locale. Pre-clinical models and clinical trials have demonstrated that in spite of their location within the central nervous system (CNS), a tissue described as 'immune privileged', brain tumors can be effectively targeted by the activated immune system following various immunotherapeutic strategies. This review will highlight recent advances in brain tumor immunotherapy, with particular emphasis on advances made using gene therapy strategies, as well as reviewing other novel therapies that can be used in combination with immunotherapy. Another important aspect of implementing gene therapy in the clinical arena is to be able to image the targeting of the therapeutics to the tumors, treatment effectiveness and progression of disease. We have therefore reviewed the most exciting non-invasive, in vivo imaging techniques which can be used in combination with gene therapy to monitor therapeutic efficacy over time.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Terapia Genética , Imunoterapia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Terapia Combinada , Progressão da Doença , Glioblastoma/diagnóstico por imagem , Glioblastoma/tratamento farmacológico , Glioblastoma/imunologia , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Interferons/uso terapêutico , Interleucinas/uso terapêutico , Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Toxinas Biológicas/uso terapêutico
17.
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
18.
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
19.
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
20.
PLoS One ; 6(8): e23523, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21897844

RESUMO

CD8(+) T cells infiltrate the brain during an anti-viral immune response. Within the brain CD8(+) T cells recognize cells expressing target antigens, become activated, and secrete IFNγ. However, there are no methods to recognize individual cells that respond to IFNγ. Using a model that studies the effects of the systemic anti-adenoviral immune response upon brain cells infected with an adenoviral vector in mice, we describe a method that identifies individual cells that respond to IFNγ. To identify individual mouse brain cells that respond to IFNγ we constructed a series of adenoviral vectors that contain a transcriptional response element that is selectively activated by IFNγ signaling, the gamma-activated site (GAS) promoter element; the GAS element drives expression of a transgene, Cre recombinase (Ad-GAS-Cre). Upon binding of IFNγ to its receptor, the intracellular signaling cascade activates the GAS promoter, which drives expression of the transgene Cre recombinase. We demonstrate that upon activation of a systemic immune response against adenovirus, CD8(+) T cells infiltrate the brain, interact with target cells, and cause an increase in the number of cells expressing Cre recombinase. This method can be used to identify, study, and eventually determine the long term fate of infected brain cells that are specifically targeted by IFNγ. The significance of this method is that it will allow to characterize the networks in the brain that respond to the specific secretion of IFNγ by anti-viral CD8(+) T cells that infiltrate the brain. This will allow novel insights into the cellular and molecular responses underlying brain immune responses.


Assuntos
Antivirais/metabolismo , Encéfalo/citologia , Encéfalo/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon gama/metabolismo , Imagem Molecular/métodos , Transdução de Sinais/imunologia , Adenoviridae/genética , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Feminino , Genes Reporter/genética , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Integrases/metabolismo , Interferon gama/genética , Masculino , Camundongos , Regiões Promotoras Genéticas/genética , Especificidade da Espécie , Timidina Quinase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA