Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bull Cancer ; 98(2): 120-32, 2011 Feb.
Artigo em Francês | MEDLINE | ID: mdl-21382793

RESUMO

Animal experimentation is a prerequisite for preclinical evaluation of treatments such as chemotherapy. It's strictly regulated with the purpose of reducing the number of experimental animal as well as their pain. Small animal imaging should provide a painless longitudinal follow up of tumor progression on a single animal. The aim of the study is to validate small animal imaging by microscanner (µscan) in longitudinal follow up of a hepatocellular carcinoma (HCC) and to demonstrate its interest for in vivo evaluation of tumor response to different therapeutics. An HCC model achieved by orthotopic graft of the MH3924A cell line in ACI rats was followed using a Imtek/Siemens microscanner (µscan) with contrast agents (Fenestra(®) LC/VC). The procedures giving the optimal enhancement of the liver as well as a reliable determination of tumor volumes by µscan were validated. Three protocols for therapeutic assessment through µscan longitudinal follow up were performed. Each consisted in three groups testing a chemotherapy (gemcitabine, gemcitabine-oxaliplatine or sorafenib) versus two control groups (placebo and doxorubicine). Comparison was done on tumor volumes, median and actual survivals. There was a significant correlation between tumor volumes measured by µscan and autopsy. Treatment by sorafenib, at the contrary of gemcitabine alone or with oxaliplatine, resulted in a significant reduction in tumor volumes and prolongation of actuarial survival. These results are consistent with available clinical data for these diverse therapeutics. In conclusion, small animal imaging with µscan is a non-invasive, reliable, and reproducible method for preclinical evaluation of antitumor agents.


Assuntos
Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/tratamento farmacológico , Microtomografia por Raio-X/métodos , Animais , Antineoplásicos/uso terapêutico , Benzenossulfonatos/administração & dosagem , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Meios de Contraste/administração & dosagem , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Modelos Animais de Doenças , Progressão da Doença , Doxorrubicina/administração & dosagem , Monitoramento de Medicamentos/métodos , Neoplasias Hepáticas/patologia , Masculino , Niacinamida/análogos & derivados , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Compostos de Fenilureia , Piridinas/administração & dosagem , Ratos , Ratos Endogâmicos ACI , Sorafenibe , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
2.
Mol Ther ; 17(7): 1164-72, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19367260

RESUMO

The incidence of lymphomas developing in both immunocompetent and immunosuppressed patients continues to steadily increase worldwide. Current chemotherapy and immunotherapy approaches have several limitations, such as severe side toxicity and selection of resistant cell variants. Autonomous parvoviruses (PVs), in particular the rat parvovirus H-1PV, have emerged as promising anticancer agents. Although it is apathogenic in humans, H-1PV has been shown to infect and suppress various rat and human tumors in animal models. In this study, we demonstrate the capacity of H-1PV for efficiently killing, through necrosis, cell cultures originating from Burkitt's lymphoma (BL), while sparing normal B lymphocytes. The cytotoxic effect was generally accompanied by a productive H-1PV infection. Remarkably, parvovirus-based monotherapy efficiently suppressed established BL at an advanced stage in a severe combined immunodeficient (SCID) mouse model of the disease. The data show for the first time that an oncolytic parvovirus deserves further consideration as a potential tool for the treatment of some non-Hodgkin B-cell lymphomas, including those resistant to apoptosis induction by rituximab.


Assuntos
Linfoma/terapia , Terapia Viral Oncolítica/métodos , Parvovirus/fisiologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Linfoma de Células B/terapia , Camundongos , Camundongos SCID , Necrose/virologia , Parvovirus/genética , Ratos , Replicação Viral/genética
3.
Clin Cancer Res ; 15(2): 511-9, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19147756

RESUMO

UNLABELLED: Pancreatic carcinoma is a gastrointestinal malignancy with poor prognosis. Treatment with gemcitabine, the most potent chemotherapeutic against this cancer up to date, is not curative, and resistance may appear. Complementary treatment with an oncolytic virus, such as the rat parvovirus H-1PV, which is infectious but nonpathogenic in humans, emerges as an innovative option. PURPOSE: To prove that combining gemcitabine and H-1PV in a model of pancreatic carcinoma may reduce the dosage of the toxic drug and/or improve the overall anticancer effect. EXPERIMENTAL DESIGN: Pancreatic tumors were implanted orthotopically in Lewis rats or subcutaneously in nude mice and treated with gemcitabine, H-1PV, or both according to different regimens. Tumor size was monitored by micro-computed tomography, whereas bone marrow, liver, and kidney functions were monitored by measuring clinically relevant markers. Human pancreatic cell lines and gemcitabine-resistant derivatives were tested in vitro for sensitivity to H-1PV infection with or without gemcitabine. RESULTS: In vitro studies proved that combining gemcitabine with H-1PV resulted in synergistic cytotoxic effects and achieved an up to 15-fold reduction in the 50% effective concentration of the drug, with drug-resistant cells remaining sensitive to virus killing. Toxicologic screening showed that H-1PV had an excellent safety profile when applied alone or in combination with gemcitabine. The benefits of applying H-1PV as a second-line treatment after gemcitabine included reduction of tumor growth, prolonged survival of the animals, and absence of metastases on CT-scans. CONCLUSION: In addition to their potential use as monotherapy for pancreatic cancer, parvoviruses can be best combined with gemcitabine in a two-step protocol.


Assuntos
Carcinoma/tratamento farmacológico , Desoxicitidina/análogos & derivados , Vírus Oncolíticos/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Parvovirus/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Carcinoma/terapia , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas/terapia , Ratos , Ratos Endogâmicos Lew , Gencitabina
4.
Oncol Rep ; 17(6): 1493-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17487410

RESUMO

Oncolytic viruses have emerged as a novel class of potent anticancer agents offering an improvement on chemo- and radiotherapy in terms of tumor targeting and reduction of side-effects. Among these agents, autonomous parvoviruses have attracted the attention of researchers for their ability to preferentially replicate in and kill transformed cells, and to suppress tumors in the absence of adverse reactions in various animal models. We have previously shown that lethally irradiated autologous tumor cells can support parvovirus H-1PV production and serve as carriers to deliver progeny H-1PV into the vicinity of lung metastases in a rat tumor model, resulting in H-1PV infection of and multiplication in metastatic cells. It is known that irradiated autologous (neoplastic) cells can also act as a therapeutic vaccine against the original tumor. Yet the ability of these cells to suppress metastases in the above model was found to be much increased as a result of their H-1PV infection. This prompted us to determine whether H-1PV boosted the tumor-suppressing capacity of the autologous vaccine by increasing its immunogenic potential and/or by making it a factory of oncolytic viruses able to reach and destroy the metastases. Both effects could be dissociated in the presence of neutralising antibodies which either prevent the progeny viruses from spreading to metastatic cells, or deplete the CD8 effector cells from the immune system. This strategy revealed that the H-1PV infection of tumor cells enhanced their ability to trigger an immune response for which uninfected tumor cells could be the targets, thereby amplifying and taking over from the direct viral oncolytic activity. This dual oncolytic/vaccinal effect of H-1PV holds out promises of clinical applications to cancer therapy.


Assuntos
Vacinas Anticâncer , Parvovirus H-1/fisiologia , Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Animais , Anticorpos Antivirais/farmacologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Parvovirus H-1/efeitos dos fármacos , Parvovirus H-1/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Vírus Oncolíticos/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Virol ; 79(6): 3517-24, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15731246

RESUMO

Due to their oncolytic properties and apathogenicity, autonomous parvoviruses have attracted significant interest as possible anticancer agents. Recent preclinical studies provided evidence of the therapeutic potential of minute virus of mice prototype strain (MVMp) and its recombinant derivatives. In a murine model of hemangiosarcoma, positive therapeutic outcome correlated with high intratumoral expression of MVMp-encoded genes in tumors and lymphoid organs, especially in tumor-draining lymph nodes. The source and relevance of this extratumoral expression, which came as a surprise because of the known fibrotropism of MVMp, remained unclear. In the present study, we investigated (i) whether the observed expression pattern occurs in different tumor models, (ii) which cell population is targeted by the virus, and (iii) the immunological consequences of this infection. Significant MVMp gene expression was detected in lymphoid tissues from infected tumor-free as well as melanoma-, lymphoma-, and hemangiosarcoma-bearing mice. This expression was especially marked in lymph nodes draining virus-injected tumors. Fluorescent in situ hybridization analysis, multicolor fluorescence-activated cell sorting, and quantitative reverse transcription-PCR revealed that MVMp was expressed in rare subpopulations of CD11b (Mac1)-positive cells displaying CD11c+ (myeloid dendritic cells [MDC]) or CD45B (B220+ [B1 lymphocytes]) markers. Apart from the late deletion of cytotoxic memory cells (CD8+ CD44+ CD62L-), this infection did not lead to significant alteration of the immunological profile of cells populating lymphoid organs. However, subtle changes were detected in the production of specific proinflammatory cytokines in lymph nodes from virus-treated animals. Considering the role of B1 lymphocytes and MDC in cancer and immunological surveillance, the specific ability of these cell types to sustain parvovirus-driven gene expression may be exploited in gene therapy protocols.


Assuntos
Linfócitos B/virologia , Células Dendríticas/virologia , Tecido Linfoide/virologia , Vírus Miúdo do Camundongo/genética , Infecções por Parvoviridae/virologia , Animais , Linfócitos B/imunologia , Antígeno CD11b/análise , Antígeno CD11c/análise , Células Dendríticas/imunologia , Feminino , Expressão Gênica , Genes Virais , Hemangiossarcoma/virologia , Antígenos Comuns de Leucócito/análise , Linfonodos/imunologia , Linfonodos/virologia , Subpopulações de Linfócitos/imunologia , Tecido Linfoide/imunologia , Linfoma/virologia , Melanoma Experimental/virologia , Camundongos , Vírus Miúdo do Camundongo/crescimento & desenvolvimento , Infecções por Parvoviridae/imunologia , Baço/imunologia , Baço/virologia , Transcrição Gênica
6.
Curr Gene Ther ; 4(3): 249-61, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15384939

RESUMO

Parvoviruses are small nuclear replicating DNA viruses. The rodent parvoviruses are usually weakly pathogenic in adult animals, bind to cell surface receptors which are fairly ubiquitously expressed on cells, and do not appear to integrate into host chromosomes during either lytic or persistent infection. The closely related rodent parvoviruses MVM, H-1 and LuIII efficiently infect human cell lines. Most interesting, malignant transformation of human and rodent cells was often found to correlate with a greater susceptibility to parvovirus-induced killing (oncolysis) and with an increase in the cellular capacity for amplifying and / or expressing the incoming parvoviral DNA. These and other interesting properties make these autonomous rodent parvoviruses and recombinant derivatives promising candidate antitumor vectors. Capsid replacement vectors have been produced from MVM or H-1 virus that carry transgenes encoding either therapeutic products (cytokines/chemokines, Apoptin, herpes simplex virus thymidine kinase) or marker proteins (green fluorescent protein, chloramphenicolacetyl transferase, luciferase). This review describes the current state of the art regarding the potential application of wild-type parvoviruses and derived vectors for the treatment of cancer. In particular, recent successes with the development of replication-competent virus-free vector stocks are discussed and results from pre-clinical studies using recombinant parvoviruses transducing various cytokines/chemokines are presented.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Neoplasias/terapia , Parvovirus/fisiologia , Replicação Viral , Humanos , Neoplasias/genética
7.
Int J Cancer ; 109(5): 742-9, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-14999784

RESUMO

Over the last few years, naturally occurring or genetically manipulated oncolytic viruses gained increasing attention as novel therapeutics for cancer treatment. The present work provides proof of principle that an organotropic cell-based carrier system is suitable to deliver oncolytic parvoviruses to a tissue known to be a target for the formation of metastases. Carrier cells were inactivated by gamma-irradiation after infection, which was found not to affect the production and release of parvoviruses that were capable of lysing cocultured target neoplastic cells. Although systemically administered parvovirus H-1 showed a pronounced therapeutic effect against the development of established Morris hepatoma (MH3924A) lung metastases, the carrier cell strategy offered a number of advantages. Infected carriers were able to sustain H-1 virus expression for 6 days in the lungs of rats affected by metastatic disease and to reduce the spreading of the virus to peripheral organs. Compared to direct virus injection, the carrier cell protocol led to an improved therapeutic effect (metastases suppression) and a lesser generation of virus-neutralizing antibodies. These data support the use of carrier cells to deliver oncolytic viruses and/or viral vectors locally in tumors and, more particularly, metastases.


Assuntos
Neoplasias Hepáticas Experimentais/terapia , Neoplasias Pulmonares/terapia , Parvovirus , Animais , Anticorpos Antineoplásicos , Anticorpos Antivirais , Técnicas de Cocultura , DNA Viral , Modelos Animais de Doenças , Estudos de Viabilidade , Imunofluorescência , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Pulmonares/secundário , Microscopia de Fluorescência , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...