Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Mol Ther ; 21(10): 1930-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23842448

RESUMO

Oncolytic viruses are structurally and biologically diverse, spreading through tumors and killing them by various mechanisms and with different kinetics. Here, we created a hybrid vesicular stomatitis/measles virus (VSV/MV) that harnesses the safety of oncolytic MV, the speed of VSV, and the tumor killing mechanisms of both viruses. Oncolytic MV targets CD46 and kills by forcing infected cells to fuse with uninfected neighbors, but propagates slowly. VSV spreads rapidly, directly lysing tumor cells, but is neurotoxic and loses oncolytic potency when neuroattenuated by conventional approaches. The hybrid VSV/MV lacks neurotoxicity, replicates rapidly with VSV kinetics, and selectively targets CD46 on tumor cells. Its in vivo performance in a myeloma xenograft model was substantially superior to either MV or widely used recombinant oncolytic VSV-M51.


Assuntos
Engenharia Genética , Vírus do Sarampo/fisiologia , Proteína Cofatora de Membrana/efeitos dos fármacos , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Células CHO , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetulus , Modelos Animais de Doenças , Humanos , Vírus do Sarampo/genética , Camundongos , Camundongos SCID , Camundongos Transgênicos , Mieloma Múltiplo/patologia , Neurônios/patologia , Neurônios/virologia , Vírus Oncolíticos/genética , Células Vero , Vírus da Estomatite Vesicular Indiana/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Transl Med ; 11: 20, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23347343

RESUMO

BACKGROUND: Mesenchymal stem cells (MSC) can serve as carriers to deliver oncolytic measles virus (MV) to ovarian tumors. In preparation for a clinical trial to use MSC as MV carriers, we obtained cells from ovarian cancer patients and evaluated feasibility and safety of this approach. METHODS: MSC from adipose tissues of healthy donors (hMSC) and nine ovarian cancer patients (ovMSC) were characterized for susceptibility to virus infection and tumor homing abilities. RESULTS: Adipose tissue (range 0.16-3.96 grams) from newly diagnosed and recurrent ovarian cancer patients yielded about 7.41×106 cells at passage 1 (range 4-9 days). Phenotype and doubling times of MSC were similar between ovarian patients and healthy controls. The time to harvest of 3.0×108 cells (clinical dose) could be achieved by day 14 (range, 9-17 days). Two of nine samples tested had an abnormal karyotype represented by trisomy 20. Despite receiving up to 1.6×109 MSC/kg, no tumors were seen in SCID beige mice and MSC did not promote the growth of SKOV3 human ovarian cancer cells in mice. The ovMSC migrated towards primary ovarian cancer samples in chemotaxis assays and to ovarian tumors in athymic mice. Using non-invasive SPECT-CT imaging, we saw rapid co-localization, within 5-8 minutes of intraperitoneal administration of MV infected MSC to the ovarian tumors. Importantly, MSC can be pre-infected with MV, stored in liquid nitrogen and thawed on the day of infusion into mice without loss of activity. MV infected MSC, but not virus alone, significantly prolonged the survival of measles immune ovarian cancer bearing animals. CONCLUSIONS: These studies confirmed the feasibility of using patient derived MSC as carriers for oncolytic MV therapy. We propose an approach where MSC from ovarian cancer patients will be expanded, frozen and validated to ensure compliance with the release criteria. On the treatment day, the cells will be thawed, washed, mixed with virus, briefly centrifuged and incubated for 2 hours with virus prior to infusion of the virus/MSC cocktail into patients.


Assuntos
Vírus do Sarampo/fisiologia , Células-Tronco Mesenquimais/citologia , Terapia Viral Oncolítica , Neoplasias Ovarianas/terapia , Animais , Estudos de Casos e Controles , Estudos de Viabilidade , Feminino , Humanos , Cariotipagem , Células-Tronco Mesenquimais/virologia , Camundongos , Camundongos Nus , Camundongos SCID , Neoplasias Ovarianas/genética , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
3.
Hum Gene Ther ; 21(4): 451-62, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19911974

RESUMO

Toxicology studies were performed in rats and rhesus macaques to establish a safe starting dose for intratumoral injection of an oncolytic vesicular stomatitis virus expressing human interferon-beta (VSV-hIFNbeta) in patients with hepatocellular carcinoma (HCC). No adverse events were observed after administration of 7.59 x 10(9) TCID(50) (50% tissue culture infective dose) of VSV-hIFNbeta into the left lateral hepatic lobe of Harlan Sprague Dawley rats. Plasma alanine aminotransferase and alkaline phosphatase levels increased and platelet counts decreased in the virus-treated animals on days 1 and 2 but returned to pretreatment levels by day 4. VSV-hIFNbeta was also injected into normal livers or an intrahepatic McA-RH7777 HCC xenograft established in Buffalo rats. Buffalo rats were more sensitive to neurotoxic effects of VSV; the no observable adverse event level (NOAEL) of VSV-hIFNbeta in Buffalo rats was 10(7) TCID(50). Higher doses were associated with fatal neurotoxicity and infectious virus was recovered from tumor and brain. Compared with VSV-hIFNbeta, toxicity of VSV-rIFNbeta (recombinant VSV expressing rat IFN-beta) was greatly diminished in Buffalo rats (NOAEL, >10(10) TCID(50)). Two groups of two adult male rhesus macaques received 10(9) or 10(10) TCID(50) of VSV-hIFNbeta injected directly into the left hepatic lobe under computed tomographic guidance. No neurological signs were observed at any time point. No abnormalities (hematology, clinical chemistry, body weights, behavior) were seen and all macaques developed neutralizing anti-VSV antibodies. Plasma interleukin-6, tumor necrosis factor-alpha, and hIFN-beta remained below detection levels by ELISA. On the basis of these studies, we will be proposing a cautious approach to dose escalation in a phase I clinical trial among patients with HCC.


Assuntos
Carcinoma Hepatocelular/virologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Interferon beta/metabolismo , Neoplasias Hepáticas Experimentais/virologia , Fígado/virologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Carcinoma Hepatocelular/terapia , Chlorocebus aethiops , Vias de Administração de Medicamentos , Feminino , Humanos , Interferon beta/genética , Fígado/patologia , Neoplasias Hepáticas Experimentais/terapia , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento , Células Vero , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/metabolismo
4.
Clin Cancer Res ; 15(23): 7246-55, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19934299

RESUMO

PURPOSE: Preexisting antiviral antibodies in cancer patients can quickly neutralize oncolytic measles virus (MV) and decrease its antitumor potency. In contrast to "naked" viruses, cell-associated viruses are protected from antibody neutralization. Hence, we hypothesized that measles virotherapy of ovarian cancer in measles-immune mice might be superior if MV-infected mesenchymal stem cell (MSC) carriers are used. EXPERIMENTAL DESIGN: Antimeasles antibodies titers in ovarian cancer patients were determined. The protection of MV by MSC from antimeasles antibodies, the in vivo biodistribution profiles, and tumor infiltration capability of MSC were determined. Measles-naïve or immune tumor-bearing mice were treated with naked virus or MSC-associated virus and mice survivals were compared. RESULTS: MSC transferred MV infection to target cells via cell-to-cell heterofusion and induced syncytia formation in the presence of high titers of antimeasles antibody, at levels that completely inactivated naked virus. Athymic mice bearing i.p. human SKOV3ip.1 ovarian tumor xenografts passively immunized with measles-immune human serum were treated with saline, naked MV, or MV-infected MSC. Bioluminescent and fluorescent imaging data indicated that i.p. administered MSC localized to peritoneal tumors, infiltrated into the tumor parenchyma, and transferred virus infection to tumors in measles naïve and passively immunized mice. Survival of the measles-immune mice was significantly enhanced by treatment with MV-infected MSC. In contrast, survivals of passively immunized mice were not prolonged by treatment with naked virus or uninfected MSC. CONCLUSIONS: MSC should be used as carriers of MV for intraperitoneal virotherapy in measles-immune ovarian cancer patients.


Assuntos
Vírus do Sarampo/metabolismo , Células-Tronco Mesenquimais/citologia , Terapia Viral Oncolítica/métodos , Neoplasias Ovarianas/terapia , Animais , Vacinas Anticâncer , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Humanos , Imunoglobulina G/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Vírus Oncolíticos/metabolismo , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA