Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 11: 464, 2011 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22029859

RESUMO

BACKGROUND: Parvovirus H-1 (H-1PV) infects and lyses human tumor cells including melanoma, hepatoma, gastric, colorectal, cervix and pancreatic cancers. We assessed whether the beneficial effects of chemotherapeutic agents or targeted agents could be combined with the oncolytic and immunostimmulatory properties of H-1PV. METHODS: Using human ex vivo models we evaluated the biological and immunological effects of H-1PV-induced tumor cell lysis alone or in combination with chemotherapeutic or targeted agents in human melanoma cells +/- characterized human cytotoxic T-cells (CTL) and HLA-A2-restricted dendritic cells (DC). RESULTS: H-1PV-infected MZ7-Mel cells showed a clear reduction in cell viability of >50%, which appeared to occur primarily through apoptosis. This correlated with viral NS1 expression levels and was enhanced by combination with chemotherapeutic agents or sunitinib. Tumor cell preparations were phagocytosed by DC whose maturation was measured according to the treatment administered. Immature DC incubated with H-1PV-induced MZ7-Mel lysates significantly increased DC maturation compared with non-infected or necrotic MZ7-Mel cells. Tumor necrosis factor-α and interleukin-6 release was clearly increased by DC incubated with H-1PV-induced SK29-Mel tumor cell lysates (TCL) and was also high with DC-CTL co-cultures incubated with H-1PV-induced TCL. Similarly, DC co-cultures with TCL incubated with H-1PV combined with cytotoxic agents or sunitinib enhanced DC maturation to a greater extent than cytotoxic agents or sunitinib alone. Again, these combinations increased pro-inflammatory responses in DC-CTL co-cultures compared with chemotherapy or sunitinib alone. CONCLUSIONS: In our human models, chemotherapeutic or targeted agents did not only interfere with the pronounced immunomodulatory properties of H-1PV, but also reinforced drug-induced tumor cell killing. H-1PV combined with cisplatin, vincristine or sunitinib induced effective immunostimulation via a pronounced DC maturation, better cytokine release and cytotoxic T-cell activation compared with agents alone. Thus, the clinical assessment of H-1PV oncolytic tumor therapy not only alone but also in combination strategies is warranted.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Parvovirus H-1/fisiologia , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Infecções por Parvoviridae/virologia , Neoplasias Cutâneas/terapia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada/métodos , Citocinas/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/virologia , Modelos Biológicos , Vírus Oncolíticos/fisiologia , Infecções por Parvoviridae/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/virologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/virologia , Células Tumorais Cultivadas/efeitos dos fármacos , Proteínas Virais/metabolismo
2.
World J Gastroenterol ; 14(24): 3819-28, 2008 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-18609705

RESUMO

AIM: To evaluate the synergistic targeting and killing of human hepatocellular carcinoma (HCC) cells lacking p53 by the oncolytic autonomous parvovirus (PV) H-1 and chemotherapeutic agents and its dependence on functional promyelocytic leukemia protein (PML). METHODS: The role of p53 and PML in regulating cytotoxicity and gene transfer mediated by wild-type (wt) PV H-1 were explored in two pairs of isogenic human hepatoma cell lines with different p53 status. Furthermore, H-1 PV infection was combined with cytostatic drug treatment. RESULTS: While the HCC cells with different p53 status studied were all susceptible to H-1 PV-induced apoptosis, the cytotoxicity of H-1 PV was more pronounced in p53-negative than in p53-positive cells. Apoptosis rates in p53-negative cell lines treated by genotoxic drugs were further enhanced by a treatment with H-1 PV. In flow cytometric analyses, H-1 PV infection resulted in a reduction of the mitochondrial transmembrane potential. In addition, H-1 PV cells showed a significant increase in PML expression. Knocking down PML expression resulted in a striking reduction of the level of H-1 PV infected tumor cell death. CONCLUSION: H-1 PV is a suitable agent to circumvent the resistance of p53-negative HCC cells to genotoxic agents, and it enhances the apoptotic process which is dependent on functional PML. Thus, H-1 PV and its oncolytic vector derivatives may be considered as therapeutic options for HCC, particularly for p53-negative tumors.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/virologia , Parvovirus H-1/patogenicidade , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/virologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Terapia Combinada , Tratamento Farmacológico , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Humanos , Neoplasias Hepáticas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteína da Leucemia Promielocítica , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo
3.
Hum Gene Ther ; 16(8): 996-1005, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16076257

RESUMO

Oncotropic and oncolytic viruses have attracted high attention as antitumor agents because they preferentially kill cancer cells in vitro and reduce the incidence of spontaneous, induced, or implanted animal tumors. Some autonomous parvoviruses (H-1, minute virus of mice) and derived recombinant vectors are currently under preclinical evaluation. Still not fully understood, their antitumor properties involve more than just tumor cell killing. Because wild-type parvovirus-mediated tumor cell lysates (TCLs) may trigger antigen-presenting cells (APCs) to augment the host immune repertoire, we analyzed phagocytosis, maturation, and crosspresentation of H-1-induced TCLs by human dendritic cells (DCs). We first established H-1-mediated oncolysis in two HLA-A2(+) and A2(-) variant melanoma cell clones. Monocyte-derived immature DCs phagocytosed H- 1-infected TCLs as well as ultraviolet-induced apoptotic TCLs and better than freeze-thaw-induced necrotic TCLs. Immature DCs incubated with H-1-induced TCLs acquired specific maturation markers comparable to a standard cytokine cocktail. Furthermore, A2(+) DCs pulsed with H-1-infected A2(-) TCLs cross-presented melanoma antigens to specific cytotoxic T lymphocytes (CTLs) and released proinflammatory cytokines. This shows for the first time that tumor cell killing by a wild-type oncolytic virus directly stimulates human APCs and CTLs. Because H-1-infected tumors enhance the immune repertoire, the clinical perspectives of parvoviral vectors are even more promising.


Assuntos
Apoptose , Células Dendríticas/fisiologia , Melanoma/patologia , Parvovirus/imunologia , Parvovirus/patogenicidade , Fagocitose , Neoplasias Cutâneas/patologia , Animais , Antígenos de Neoplasias/imunologia , Diferenciação Celular , Apresentação Cruzada , Criopreservação , Células Dendríticas/imunologia , Antígeno HLA-A2/análise , Humanos , Camundongos , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
4.
Cancer Gene Ther ; 10(6): 477-80, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12768193

RESUMO

Certain autonomous parvoviruses preferentially replicate in and kill in vitro-transformed cells and may reduce the incidence of spontaneous and implanted tumors in animals. Hence, these viruses and their derivatives are currently under evaluation as antitumor vectors. However, the mechanisms underlying their tumor-suppressing properties are not yet understood. We asked whether the lytic parvovirus H1 may enhance the immunogenicity of infected tumor cells. Out of human melanoma and gastrointestinal tumor cells, we selected the cell line SK29-Mel-1 being very susceptible to H1-induced apoptotic killing. Here, no upregulation of HLA class I and costimulatory molecules could be observed following H1 infection. However, a strong release of the immunogenic signal-the inducible heat-shock protein HSP72, but not constitutive HSP73-was observed after H1 infection. The HSP72 release was higher and of longer duration than a conventional heat-shock treatment. We also explored H1 replication and cytotoxicity in human immune cells, as such cells may constitute targets for H1 virus replication. Long-term cultured lymphocytes, monocytes, immature and mature dendritic cells were not susceptible to H1 virus. Altogether, parvovirus-mediated cell killing may in vivo enhance tumor immunogenicity by HSP72 release and thus contribute to the antitumor effect of parvoviruses.


Assuntos
Vetores Genéticos/genética , Proteínas de Choque Térmico HSP70 , Proteínas de Choque Térmico/metabolismo , Parvovirus/genética , Apoptose , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Células Cultivadas , Proteínas de Choque Térmico HSC70 , Proteínas de Choque Térmico HSP72 , Humanos , Melanoma/metabolismo , Melanoma/terapia , Fatores de Tempo , Transgenes , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...