Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Lett ; 428: 139-146, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29709703

RESUMO

Over the last decade, autophagy has emerged as one of the critical cellular systems that control homeostasis. Besides management of normal homeostatic processes, autophagy can also be induced by tissue damage stress or by rapidly progressing tumors. During tumor progression, autophagy mediates a cellular reaction to the changes inside and outside of cells, which leads to tumor adaptation. Even though the regulation of autophagy seems universal and is a well-described process, its dysregulation and role in glioma progression remain an important topic of investigation. In this review, we summarize recent evidence of autophagy regulation in brain tumor tissues and possible interconnection between signaling pathways that govern cellular responses. This perspective may help to assess the qualitative differences and various outcomes in response to autophagy stimulation.


Assuntos
Proteínas Relacionadas à Autofagia/genética , Autofagia/genética , Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Proteínas Relacionadas à Autofagia/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cloroquina/farmacologia , Cloroquina/uso terapêutico , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Glioma/mortalidade , Glioma/patologia , Glioma/terapia , Humanos , MicroRNAs/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Terapia Viral Oncolítica , RNA Longo não Codificante/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
2.
Cancer Gene Ther ; 21(1): 38-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24434573

RESUMO

Myeloid-derived suppressor cells (MDSCs) accumulate in the glioma microenvironment during tumor progression and promote immunosuppression. Interleukin-12 (IL-12) immunogene therapy can alter MDSCs toward an antigen-presenting cell phenotype and these mature cells can have a central role in antigen presentation. It remains unclear, however, how MDSC depletion can affect glioma immunotherapy. In this study, we generated a replication-deficient adenoviral vector, Ad.5/3.cRGD-mIL12p70, that transduces the GL261-based murine glioma cell line, resulting in the induction of biologically active, murine IL12p70 expression. Ex vivo, IL-12 expressed by GL261 cells induced interferon-γ synthesis in CD8(+) T cells (P<0.001), CD4(+) T cells (P=0.009) and natural killer cells (P=0.036). When injected 1 week after tumor implantation, Ad.5/3.cRGD-mIL12p70 successfully prolonged the survival of glioma-bearing mice. Sixty percent of animals treated with IL-12 immunotherapy were long-term survivors over 175 days, whereas all the control group animals expired by 40 days after tumor implantation (P=0.026). Mice receiving Ad.5/3.cRGD-mIL12p70 also accumulated 50% less MDSCs in the brain than the control group (P=0.007). Moreover, in the IL-12 group, MDSCs significantly overexpressed CD80 and major histocompatibility complex class II molecules (P=0.041). Depletion of MDSCs with Gr1(+) antibody had no survival benefit induced by IL-12-mediated immunotherapy. Of note, IL-12 therapy increased the presence of myeloid dendritic cells (mDCs) in the glioma microenvironment (P=0.0069). Ultimately, the data show that in the context of IL-12 immunogene therapy, MDSCs are dispensable and mDCs may provide the majority of antigen presentation in the brain.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Glioma/genética , Glioma/imunologia , Interleucina-12/genética , Células Mieloides/imunologia , Adenoviridae/genética , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glioma/mortalidade , Glioma/terapia , Imunoterapia , Interleucina-12/biossíntese , Masculino , Camundongos
3.
Genes Dis ; 1(2): 214-226, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25685829

RESUMO

Glioblastoma Multiforme (GBM) is a rapidly progressing brain tumor. Despite the relatively low percentage of cancer patients with glioma diagnoses, recent statistics indicate that the number of glioma patients may have increased over the past decade. Current therapeutic options for glioma patients include tumor resection, chemotherapy, and concomitant radiation therapy with an average survival of approximately 16 months. The rapid progression of gliomas has spurred the development of novel treatment options, such as cancer gene therapy and oncolytic virotherapy. Preclinical testing of oncolytic adenoviruses using glioma models revealed both positive and negative sides of the virotherapy approach. Here we present a detailed overview of the glioma virotherapy field and discuss auxiliary therapeutic strategies with the potential for augmenting clinical efficacy of GBM virotherapy treatment.

4.
Gene Ther ; 20(3): 318-27, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22673390

RESUMO

Conditionally replicating adenoviruses (CRAd) are a promising class of gene therapy agents that can overcome already known glioblastoma (GBM) resistance mechanisms but have limited distribution upon direct intratumoral (i.t.) injection. Collagen bundles in the extracellular matrix (ECM) have an important role in inhibiting virus distribution. In fact, ECM pre-treatment with collagenases improves virus distributions to tumor cells. Matrix metalloproteinases (MMPs) are an endogenous class of collagenases secreted by tumor cells whose function can be altered by different drugs including anti-angiogenic agents, such as bevacizumab. In this study we hypothesized that upregulation of MMP activity during anti-angiogenic therapy can improve CRAd-S-pk7 distribution in GBM. We find that MMP-2 activity in human U251 GBM xenografts increases (*P=0.03) and collagen IV content decreases (*P=0.01) during vascular endothelial growth factor (VEGF-A) antibody neutralization. After proving that collagen IV inhibits CRAd-S-pk7 distribution in U251 xenografts (Spearman rho=-0.38; **P=0.003), we show that VEGF-blocking antibody treatment followed by CRAd-S-pk7 i.t. injection reduces U251 tumor growth more than each individual agent alone (***P<0.0001). Our data propose a novel approach to improve virus distribution in tumors by relying on the early effects of anti-angiogenic therapy.


Assuntos
Adenoviridae/fisiologia , Inibidores da Angiogênese/farmacologia , Colágeno/metabolismo , Glioma/terapia , Ensaios Antitumorais Modelo de Xenoenxerto , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Bloqueadores/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glioma/genética , Glioma/patologia , Humanos , Proteínas Inibidoras de Apoptose/genética , Injeções Intralesionais , Estimativa de Kaplan-Meier , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Nus , Terapia Viral Oncolítica/métodos , Polilisina/genética , Polilisina/metabolismo , Regiões Promotoras Genéticas/genética , Proteólise , Survivina , Fator A de Crescimento do Endotélio Vascular/imunologia , Replicação Viral/efeitos dos fármacos
5.
Cancer Gene Ther ; 19(6): 431-42, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22555507

RESUMO

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


Assuntos
Adenoviridae/fisiologia , Neoplasias Encefálicas/terapia , Glioma/terapia , Células-Tronco Neurais/transplante , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Adenoviridae/genética , Proteínas E1A de Adenovirus/biossíntese , Proteínas E1A de Adenovirus/genética , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Sobrevivência Celular , Cricetinae , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/biossíntese , Humanos , Luciferases de Vaga-Lume/biossíntese , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Vírus Oncolíticos/genética , Organismos Geneticamente Modificados , Proteínas Recombinantes/biossíntese , Sigmodontinae , Carga Viral , Replicação Viral
6.
Int J Nanosci ; 10(4): 899, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23105163

RESUMO

Functional nanoscale materials that possess specific physical or chemical properties can leverage energy transduction in vivo. Once these materials integrate with biomolecules they combine physical properties of inorganic material and the biorecognition capabilities of bio-organic moieties. Such nano-bio hybrids can be interfaced with living cells, the elementary functional units of life. These nano-bio systems are capable of bio-manipulation or actuation via altering intracellular biochemical pathways. Thus, nano-bio conjugates are appealing for a wide range of applications from the life sciences and nanomedicine to catalysis and clean energy production. Here we highlight recent progress in our efforts to develop smart nano-bio hybrid materials, and to study their performance within cellular machinery under application of external stimuli, such as light or magnetic fields.

7.
Br J Cancer ; 100(7): 1154-64, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19277041

RESUMO

Conditionally replicative adenoviruses (CRAds) represent a novel treatment strategy for malignant glioma. Recent studies suggest that the cytopathic effect elicited by these vectors is mediated through autophagy, a form of programmed cell death. Likewise, temozolomide (TMZ), a chemotherapeutic agent used for the treatment of malignant gliomas, also triggers autophagic cell death. In this study, we examined the potential to combine the two treatments in the setting of experimental glioma. In vitro, pretreatment with TMZ followed by CRAd-Surivin-pk7 enhanced cytotoxicity against a panel of glioma cell lines. Western blot analysis showed increased expression of BAX and p53, decreased expression of BCL2 and elevated level of APG5. Treatment with TMZ followed by CRAd-Survivin-pk7 (CRAd-S-pk7) led to a significant over-expression of autophagy markers, acidic vesicular organelles and light-chain 3 (LC3). These results were further evaluated in vivo, in which 90% of the mice with intracranial tumours were long-term survivors (>100 days) after treatment with TMZ and CRAd-S-pk7 (P<0.01). Analysis of tumours ex vivo showed expression of both LC3 and cleaved Caspase-3, proving that both autophagy and apoptosis are responsible for cell death in vivo. These results suggest that combination of chemovirotherapy offers a powerful tool against malignant glioma and should be further explored in the clinical setting.


Assuntos
Adenoviridae/fisiologia , Antineoplásicos Alquilantes/uso terapêutico , Apoptose , Autofagia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioma/terapia , Terapia Viral Oncolítica , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Terapia Combinada , Dacarbazina/uso terapêutico , Feminino , Glioma/mortalidade , Glioma/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Associadas aos Microtúbulos/análise , Mitose , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer Gene Ther ; 16(4): 362-72, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19011597

RESUMO

Conditionally replicative adenoviruses (CRAds) are often evaluated in mice; however, normal and cancerous mouse tissues are poorly permissive for human CRAds. As the cotton rat (CR) is a semipermissive animal and the Syrian hamster (SH) is a fully permissive model for adenoviral replication, we compared them in a single study following intracranial (i.c.) injection of a novel glioma-targeting CRAd. Viral genomic copies were quantified by real-time PCR in brain, blood, liver and lung. The studies were corroborated by immunohistochemical, serological and immunological assays. CR had a multiple log higher susceptibility for adenoviral infection than SH. A similar amount of genomic copies of CRAd-Survivin-pk7 and human adenovirus serotype 5 (AdWT) was found in the brain of CR and in all organs from SH. In blood and lung of CR, AdWT had more genomic copies than CRAd-Survivin-pk7 in some of the time points studied. Viral antigens were confirmed in brain slices, an elevation of serum transaminases was observed in both models, and an increase in anti-adenoviral antibodies was detected in SH sera. In conclusion, CR represents a sensitive model for studying biodistribution of CRAds after i.c. delivery, allowing for the detection of differences in the replication of CRAd-Survivin-pk7 and AdWT that were not evident in SH.


Assuntos
Adenoviridae/fisiologia , Vetores Genéticos , Vírus Oncolíticos/fisiologia , Replicação Viral , Adenoviridae/genética , Animais , Formação de Anticorpos/efeitos dos fármacos , Encéfalo/virologia , Neoplasias do Sistema Nervoso Central/terapia , Cricetinae , DNA Viral/sangue , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Glioma/terapia , Humanos , Fígado/virologia , Pulmão/virologia , Masculino , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Ratos , Especificidade da Espécie , Transaminases/sangue
9.
Gene Ther ; 16(2): 262-78, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19078993

RESUMO

Adenoviral oncolytic virotherapy represents an attractive treatment modality for central nervous system (CNS) neoplasms. However, successful application of virotherapy in clinical trials has been hampered by inadequate distribution of oncolytic vectors. Neural stem cells (NSCs) have been shown as suitable vehicles for gene delivery because they track tumor foci. In this study, we evaluated the capability of NSCs to deliver a conditionally replicating adenovirus (CRAd) to glioma. We examined NSC specificity with respect to viral transduction, migration and capacity to deliver a CRAd to tumor cells. Fluorescence-activated cell sorter (FACS) analysis of NSC shows that these cells express a variety of surface receptors that make them amenable to entry by recombinant adenoviruses. Luciferase assays with replication-deficient vectors possessing a variety of transductional modifications targeted to these receptors confirm these results. Real-time PCR analysis of the replication profiles of different CRAds in NSCs and a representative glioma cell line, U87MG, identified the CRAd-Survivin (S)-pk7 virus as optimal vector for further delivery studies. Using in vitro and in vivo migration studies, we show that NSCs infected with CRAd-S-pk7 virus migrate and preferentially deliver CRAd to U87MG glioma. These results suggest that NSCs mediate an enhanced intratumoral distribution of an oncolytic vector in malignant glioma when compared with virus injection alone.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Neurônios/virologia , Terapia Viral Oncolítica/métodos , Transplante de Células-Tronco/métodos , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Neoplasias Encefálicas/virologia , Movimento Celular/fisiologia , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/farmacocinética , Glioma/virologia , Humanos , Proteínas Inibidoras de Apoptose , Masculino , Camundongos , Camundongos Nus , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Transplante de Neoplasias , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , RNA Mensageiro/genética , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Células-Tronco/virologia , Survivina , Transcrição Gênica , Células Tumorais Cultivadas , Replicação Viral
10.
Cancer Biol Ther ; 7(5): 786-93, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18756624

RESUMO

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


Assuntos
Adenoviridae/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Animais , Linhagem Celular Tumoral , Receptor Constitutivo de Androstano , Citomegalovirus/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Humanos , Proteína Cofatora de Membrana/metabolismo , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Vírus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...