Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Head Neck ; 38(2): 237-46, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25244076

RESUMEN

BACKGROUND: Oncolytic viral efficacy may be limited by the penetration of the virus into tumors. This may be enhanced by intraoperative application of virus immediately after surgical resection. METHODS: Oncolytic vaccinia virus GLV-1h68 was delivered in silk-elastin-like protein polymer (SELP) in vitro and in vivo in anaplastic thyroid carcinoma cell line 8505c in nude mice. RESULTS: GLV-1h68 in SELP infected and lysed anaplastic thyroid cancer cells in vitro equally as effectively as in phosphate-buffered saline (PBS), and at 1 week retains a thousand fold greater infectious plaque-forming units. In surgical resection models of residual tumor, GLV-1h68 in SELP improves tumor control and shows increased viral ß-galactosidase expression as compared to PBS. CONCLUSION: The use of SELP matrix for intraoperative oncolytic viral delivery protects infectious viral particles from degradation, facilitates sustained viral delivery and transgene expression, and improves tumor control. Such optimization of methods of oncolytic viral delivery may enhance therapeutic outcomes.


Asunto(s)
Viroterapia Oncolítica/métodos , Virus Oncolíticos , Neoplasias de la Tiroides/inmunología , Virus Vaccinia , Animales , Biopolímeros , Línea Celular Tumoral , Fibroínas , Fibronectinas , Cuidados Intraoperatorios , Ratones Desnudos , Proteínas Recombinantes de Fusión , Neoplasias de la Tiroides/cirugía , Ensayo de Placa Viral , Ensayos Antitumor por Modelo de Xenoinjerto , beta-Galactosidasa/metabolismo
2.
Surgery ; 158(2): 331-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26049609

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) commonly presents at a late stage when surgery is no longer a curative option. As such, novel therapies for advanced HCC are needed. Oncolytic viruses are a viable option for cancer therapy owing to their ability to specifically infect, replicate within, and kill cancer cells. In this study, we have investigated the ability of GLV-2b372, a novel light-emitting recombinant vaccinia virus derived from a wild-type Lister strain, to kill HCC. METHODS: Four human HCC cell lines were assayed in vitro for infectivity and cytotoxicity. Viral replication was quantified via standard viral plaque assays. Flank HCC xenografts generated in athymic nude mice were treated with intratumoral GLV-2b372 to assess for tumor growth inhibition and viral biodistribution. RESULTS: Infectivity occurred in a time- and concentration-dependent manner with 70% cell death in all cell lines by day 5. All cell lines supported efficient viral replication. At 25 days after infection, flank tumor volumes decreased by 50% whereas controls increased by 400%. Tumor tissue demonstrated substantial GLV-2b372 infection at 24 hours, 48 hours, and 2 weeks. CONCLUSION: We demonstrate that GLV-2b372 efficiently kills human HCC in vitro and in vivo and is a viable treatment option for patients with HCC.


Asunto(s)
Carcinoma Hepatocelular/terapia , Terapia Genética , Neoplasias Hepáticas/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Virus Vaccinia/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
3.
PLoS One ; 9(8): e104337, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25093734

RESUMEN

Virotherapy on the basis of oncolytic vaccinia virus (VACV) infection is a promising approach for cancer therapy. In this study we describe the establishment of a new preclinical model of feline mammary carcinoma (FMC) using a recently established cancer cell line, DT09/06. In addition, we evaluated a recombinant vaccinia virus strain, GLV-5b451, expressing the anti-vascular endothelial growth factor (VEGF) single-chain antibody (scAb) GLAF-2 as an oncolytic agent against FMC. Cell culture data demonstrate that GLV-5b451 virus efficiently infected, replicated in and destroyed DT09/06 cancer cells. In the selected xenografts of FMC, a single systemic administration of GLV-5b451 led to significant inhibition of tumor growth in comparison to untreated tumor-bearing mice. Furthermore, tumor-specific virus infection led to overproduction of functional scAb GLAF-2, which caused drastic reduction of intratumoral VEGF levels and inhibition of angiogenesis. In summary, here we have shown, for the first time, that the vaccinia virus strains and especially GLV-5b451 have great potential for effective treatment of FMC in animal model.


Asunto(s)
Vectores Genéticos/genética , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Virus Oncolíticos/genética , Virus Vaccinia/genética , Animales , Gatos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Chlorocebus aethiops , Femenino , Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacocinética , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , Viroterapia Oncolítica , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/metabolismo , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Surgery ; 156(2): 263-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24957667

RESUMEN

BACKGROUND: Sorafenib is the standard systemic therapy for unresectable or recurrent hepatocellular carcinoma (HCC) but adds minimal increase in survival. Therefore, there is a great need to develop novel therapies for advanced or recurrent HCC. One emerging field of cancer treatment involves oncolytic viruses that specifically infect, replicate within, and kill cancer cells. In this study, we examined the ability of GLV-1h68, a recombinant vaccinia virus derived from the vaccine strain that was used to eradicate smallpox, to kill sorafenib-resistant (SR) HCC cell lines. METHODS: Four SR HCC cell lines were generated by repeated passage in the presence of sorafenib. Median inhibitory concentration was determined for all cell lines. The infectivity, viral replication, and cytotoxicity of GLV-1h68 were assayed for both parental and SR HCC cells. RESULTS: Infectivity increased in a time and concentration-dependent manner in all cell lines. All cell lines supported efficient replication of virus. No difference between the rates of cell death between the parental and SR cell lines was observed. CONCLUSION: Our results demonstrate that the oncolytic vaccinia virus GLV-1h68 kills both parental and SR HCC cell lines efficiently. This study indicates that patients who have failed treatment with sorafenib remain viable candidates for oncolytic therapy.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Niacinamida/análogos & derivados , Viroterapia Oncolítica/métodos , Compuestos de Fenilurea/farmacología , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Ingeniería Genética , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Niacinamida/farmacología , Sorafenib , Virus Vaccinia/fisiología , Ensayo de Placa Viral , Replicación Viral
5.
PLoS One ; 8(9): e71105, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24019862

RESUMEN

Metastatic tumor cells in body fluids are important targets for treatment, and critical surrogate markers for evaluating cancer prognosis and therapeutic response. Here we report, for the first time, that live metastatic tumor cells in blood samples from mice bearing human tumor xenografts and in blood and cerebrospinal fluid samples from patients with cancer were successfully detected using a tumor cell-specific recombinant vaccinia virus (VACV). In contrast to the FDA-approved CellSearch system, VACV detects circulating tumor cells (CTCs) in a cancer biomarker-independent manner, thus, free of any bias related to the use of antibodies, and can be potentially a universal system for detection of live CTCs of any tumor type, not limited to CTCs of epithelial origin. Furthermore, we demonstrate for the first time that VACV was effective in preventing and reducing circulating tumor cells in mice bearing human tumor xenografts. Importantly, a single intra-peritoneal delivery of VACV resulted in a dramatic decline in the number of tumor cells in the ascitic fluid from a patient with gastric cancer. Taken together, these results suggest VACV to be a useful tool for quantitative detection of live tumor cells in liquid biopsies as well as a potentially effective treatment for reducing or eliminating live tumor cells in body fluids of patients with metastatic disease.


Asunto(s)
Líquidos Corporales , Metástasis de la Neoplasia/terapia , Viroterapia Oncolítica , Virus Vaccinia/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Xenoinjertos , Humanos , Masculino , Ratones , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia
6.
Mol Ther ; 21(11): 2054-62, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23765443

RESUMEN

Recombinant human erythropoietin (rhEPO), a glycoprotein hormone regulating red blood cell (RBC) formation, is used for the treatment of cancer-related anemia. The effect of rhEPO on tumor growth, however, remains controversial. Here, we report the construction and characterization of the recombinant vaccinia virus (VACV) GLV-1h210, expressing hEPO. GLV-1h210 was shown to replicate in and kill A549 lung cancer cells in culture efficiently. In mice bearing A549 lung cancer xenografts, treatment with a single intravenous dose of GLV-1h210 resulted in tumor-specific production and secretion of functional hEPO, which exerted an effect on RBC progenitors and precursors in the mouse bone marrow, leading to a significant increase in the number of RBCs and in the level of hemoglobin. Furthermore, virally expressed hEPO, but not exogenously added rhEPO, enhanced virus-mediated green fluorescent protein (GFP) expression in tumors and subsequently accelerated tumor regression when compared with the treatment with the parental virus GLV-1h68 or GLV-1h209 that expressed a nonfunctional hEPO protein. Moreover, intratumorally expressed hEPO caused enlarged tumoral microvessels, likely facilitating virus spreading. Taken together, VACV-mediated intratumorally expressed hEPO not only enhanced oncolytic virotherapy but also simultaneously alleviated cancer-related anemia.


Asunto(s)
Anemia/terapia , Eritropoyetina/metabolismo , Neoplasias Pulmonares/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Vaccinia/genética , Anemia/complicaciones , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Eritropoyetina/genética , Proteínas Fluorescentes Verdes , Humanos , Neoplasias Hepáticas Experimentales , Masculino , Ratones , Ratones Desnudos , Microvasos/metabolismo , Virus Oncolíticos/metabolismo , Proteínas Recombinantes/metabolismo , Virus Vaccinia/metabolismo , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int J Cancer ; 133(12): 2989-99, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23729266

RESUMEN

Oncolytic viruses are currently in clinical trials for a variety of tumors, including high grade gliomas. A characteristic feature of high grade gliomas is their high vascularity and treatment approaches targeting tumor endothelium are under investigation, including bevacizumab. The aim of this study was to improve oncolytic viral therapy by combining it with ionizing radiation and to radiosensitize tumor vasculature through a viral encoded anti-angiogenic payload. Here, we show how vaccinia virus-mediated expression of a single-chain antibody targeting VEGF resulted in radiosensitization of the tumor-associated vasculature. Cell culture experiments demonstrated that purified vaccinia virus encoded antibody targeting VEGF reversed VEGF-induced radioresistance specifically in endothelial cells but not tumor cells. In a subcutaneous model of U-87 glioma, systemically administered oncolytic vaccinia virus expressing anti-VEGF antibody (GLV-1h164) in combination with fractionated irradiation resulted in enhanced tumor growth inhibition when compared to nonanti-VEGF expressing oncolytic virus (GLV-1h68) and irradiation. Irradiation of tumor xenografts resulted in an increase in VACV replication of both GLV-1h68 and GLV-1h164. However, GLV-1h164 in combination with irradiation resulted in a drastic decrease in intratumoral VEGF levels and tumor vessel numbers in comparison to GLV-1h68 and irradiation. These findings demonstrate the incorporation of an oncolytic virus expressing an anti-VEGF antibody (GLV-1h164) into a fractionated radiation scheme to target tumor cells by enhanced VACV replication in irradiated tumors as well as to radiosensitize tumor endothelium which results in enhanced efficacy of combination therapy of human glioma xenografts.


Asunto(s)
Endotelio Vascular/efectos de la radiación , Glioma/terapia , Viroterapia Oncolítica/métodos , Tolerancia a Radiación , Virus Vaccinia/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Glioma/irrigación sanguínea , Humanos , Masculino , Ratones , Factor A de Crecimiento Endotelial Vascular/fisiología
8.
JAMA Otolaryngol Head Neck Surg ; 139(2): 173-82, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23429949

RESUMEN

OBJECTIVE: To examine the therapeutic effects of an attenuated, replication-competent vaccinia virus (GLV-1h68) against a panel of 5 human salivary gland carcinoma cell lines. DESIGN: The susceptibility of 5 salivary gland carcinoma cell lines to infection and oncolysis by GLV-1h68 was assessed in vitro and in vivo. RESULTS: All 5 cell lines were susceptible to viral infection, transgene expression, and cytotoxic reactions. Three cell lines were exquisitely sensitive to infection by very low doses of GLV-1h68. Orthotopic parotid tumors exhibited more aggressive behavior compared with flank tumors. A single intratumoral injection of GLV-1h68 induced significant tumor regression without observed toxic effects in flank and parotid tumor models; controls demonstrated rapid tumor progression. CONCLUSION: These promising results demonstrate significant oncolytic activity by an attenuated vaccinia virus for infecting and lysing salivary gland carcinomas, supporting future clinical trials.


Asunto(s)
Adenocarcinoma/terapia , Carcinoma Mucoepidermoide/terapia , Viroterapia Oncolítica , Neoplasias de las Glándulas Salivales/terapia , Virus Vaccinia/fisiología , Adenocarcinoma/patología , Animales , Carcinoma Mucoepidermoide/patología , Línea Celular Tumoral , Expresión Génica , Humanos , Inyecciones , Luciferasas/genética , Ratones , Ratones Desnudos , Neoplasias de las Glándulas Salivales/patología , Virus Vaccinia/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , beta-Galactosidasa/genética
9.
Bioengineered ; 4(2): 84-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23093804

RESUMEN

Virotherapy on the basis of oncolytic vaccinia virus (VACV) strains is one novel approach for canine cancer therapy. In this study we described for the first time the characterization and the use of new VACV strain LIVP6.1.1 as an oncolytic agent against canine cancer in a panel of four canine cancer cell lines including: soft tissue sarcoma (STSA-1), melanoma (CHAS), osteosarcoma (D-17) and prostate carcinoma (DT08/40). Cell culture data demonstrated that LIVP6.1.1 efficiently infected and destroyed all four tested canine cancer cell lines. In two different xenograft models on the basis of the canine soft tissue sarcoma STSA-1 and the prostate carcinoma DT08/40 cell lines, a systemic administration of the LIVP6.1.1 virus was found to be safe and led to anti-tumor and immunological effects resulting in the significant reduction of tumor growth in comparison to untreated control mice. In summary, the pre-clinical evaluation has demonstrated the efficacy of LIVP6.1.1 for canine cancer therapy. Furthermore, a clinical trial with canine cancer patients has already been started.


Asunto(s)
Neoplasias/terapia , Virus Oncolíticos/fisiología , Virus Vaccinia/fisiología , Animales , Perros , Ratones , Neoplasias/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Vaccinia/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Hum Gene Ther Methods ; 23(5): 285-96, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23131031

RESUMEN

Vaccinia virus (VACV) has emerged as an attractive tool in oncolytic virotherapy. VACV replication efficiency plays a crucial role in the therapeutic outcome. However, little is known about the influence of host factors on viral replication efficiency and permissiveness of a host cell line to infection and oncolysis. In this study, replication of the attenuated VACV GLV-1h68 strain and three wild-type VACV isolates was determined in two autologous human melanoma cell lines (888-MEL and 1936-MEL). Host gene expression and viral gene expression in infected cells were evaluated via respective expression array platforms. Microarray analyses followed by sequential statistical approaches characterized human genes that change specifically due to virus infection. Viral gene transcription correlated with viral replication in a time-dependent manner. A set of human genes revealed strong correlations with the respective viral gene expression. Finally we identified a set of human genes with possible predictive value for viral replication in an independent dataset. The results demonstrate a probable correlation between viral replication, early gene expression, and the respective host response, and thus a possible involvement of human host factors in viral early replication. The characterization of human target genes that influence viral replication could help answer the question of host cell permissiveness to oncolytic virotherapy and provide important information for the development of novel recombinant vaccinia viruses with improved features to enhance replication rate and hence trigger therapeutic outcome.


Asunto(s)
Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Virus Oncolíticos/fisiología , Transcripción Genética , Virus Vaccinia/fisiología , Replicación Viral , Línea Celular , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Transducción de Señal
11.
J Nucl Med ; 53(12): 1933-42, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23139088

RESUMEN

UNLABELLED: To assess therapeutic response and potential toxicity of oncolytic virotherapy, a noninvasive, deep-tissue imaging modality is needed. This study aimed to assess the feasibility, parameters, and determining factors of serial imaging and long-term monitoring of virotherapy and the therapeutic response of pancreatic cancer xenografts treated with a vaccinia virus carrying the human sodium iodide symporter GLV-1h153. METHODS: Pancreatic cancer xenografts (PANC-1) in nude mice were treated systemically or intratumorally with GLV-1h153 and serially imaged using (124)I PET at 1, 2, 3, and 5 wk after viral injection. Signal intensity was compared with tumor therapeutic response and optical imaging, and tumors were histologically analyzed for morphology and the presence of virus. Autoradiography was performed using technetium-pertechnetate and γ-scintigraphy to assess determining factors for radiouptake in tumors. Finally, the enhanced therapeutic effect of combination therapy with GLV-1h153 and systemic radioiodine was assessed. RESULTS: GLV-1h153 successfully facilitated serial long-term imaging of virotherapy, with PET signal intensity correlating to tumor response. GLV-1h153 colonization of tumors mediated radioiodine uptake at potentially therapeutic doses. Successful radiouptake required the presence of virus, adequate blood flow, and viable tissue, whereas loss of signal intensity was linked to tumor death and necrosis. Finally, combining systemically administered GLV-1h153 and (131)I led to enhanced tumor kill when compared with virus or (131)I alone (P < 0.01). CONCLUSION: GLV-1h153 is a promising oncolytic agent for the treatment, long-term imaging, and monitoring of therapeutic response in a xenograft model of pancreatic cancer. GLV-1h153 provided insight into tumor biologic activity and facilitated enhanced tumor kill when combined with systemic targeted radiotherapy. These results warrant further investigation into parameters and potential synergistic effects of combination therapy.


Asunto(s)
Imagen Molecular/métodos , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Radioterapia Guiada por Imagen/métodos , Simportadores/genética , Virus Vaccinia/genética , Animales , Línea Celular Tumoral , Estudios de Factibilidad , Humanos , Radioisótopos de Yodo , Ratones , Imagen Óptica , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Tomografía de Emisión de Positrones , Factores de Tiempo , Resultado del Tratamiento , Carga Tumoral
12.
Methods Mol Biol ; 872: 159-75, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22700410

RESUMEN

Early detection of cancer and metastases is pivotal to the success of subsequent treatment intervention. In recent years, the use of live microorganisms, such as viruses and bacteria, has gained substantial research and clinical interest in both detection and therapy of cancer. Many of these live microorganisms have shown remarkable tumor-specific replication following systemic delivery. With the aid of modern molecular technologies, modified live microorganisms can be engineered to carry additional diagnostic and therapeutic capabilities. We have shown that when armed with light-emitting protein genes, such as genes for luciferase and green fluorescent protein, the entry and specific amplification of systemically-delivered vaccinia virus and bacteria in tumors can be visualized in real time using a low-light imager, or using macro- and micro-fluorescence microscopes. Therefore, through optical imaging, the location of tumors and metastases could be revealed by these light-emitting microorganisms. The tumor-colonization capability has been demonstrated in both immuno-competent as well as immuno-compromised rodent models with syngeneic and allogeneic tumors. Based on their "tumor-finding" nature, bacteria and viruses could be further designed as "vehicles" to carry multiple genes for detection and therapy of cancer.


Asunto(s)
Diagnóstico por Imagen/métodos , Proteínas Luminiscentes/metabolismo , Animales , Bacterias/metabolismo , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Fluorescente , Neoplasias , Ratas , Virus Vaccinia/metabolismo
13.
PLoS One ; 7(5): e37239, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615950

RESUMEN

Virotherapy using oncolytic vaccinia virus (VACV) strains is one promising new strategy for canine cancer therapy. In this study we describe the establishment of an in vivo model of canine soft tissue sarcoma (CSTS) using the new isolated cell line STSA-1 and the analysis of the virus-mediated oncolytic and immunological effects of two different Lister VACV LIVP1.1.1 and GLV-1h68 strains against CSTS. Cell culture data demonstrated that both tested VACV strains efficiently infected and destroyed cells of the canine soft tissue sarcoma line STSA-1. In addition, in our new canine sarcoma tumor xenograft mouse model, systemic administration of LIVP1.1.1 or GLV-1h68 viruses led to significant inhibition of tumor growth compared to control mice. Furthermore, LIVP1.1.1 mediated therapy resulted in almost complete tumor regression and resulted in long-term survival of sarcoma-bearing mice. The replication of the tested VACV strains in tumor tissues led to strong oncolytic effects accompanied by an intense intratumoral infiltration of host immune cells, mainly neutrophils. These findings suggest that the direct viral oncolysis of tumor cells and the virus-dependent activation of tumor-associated host immune cells could be crucial parts of anti-tumor mechanism in STSA-1 xenografts. In summary, the data showed that both tested vaccinia virus strains and especially LIVP1.1.1 have great potential for effective treatment of CSTS.


Asunto(s)
Neoplasias de los Músculos/veterinaria , Viroterapia Oncolítica/métodos , Sarcoma/veterinaria , Neoplasias de los Tejidos Blandos/veterinaria , Virus Vaccinia/fisiología , Animales , Neoplasias Óseas/secundario , Neoplasias Óseas/veterinaria , Línea Celular Tumoral , Perros , Ratones , Neoplasias de los Músculos/terapia , Virus Oncolíticos/fisiología , Sarcoma/patología , Sarcoma/terapia , Neoplasias de los Tejidos Blandos/terapia , Replicación Viral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Clin Cancer Res ; 18(9): 2579-90, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22379115

RESUMEN

PURPOSE: Radiotherapy is part of the standard of care in high-grade gliomas but its outcomes remain poor. Integrating oncolytic viruses with standard anticancer therapies is an area of active investigation. The aim of this study was to determine how tumor-targeted ionizing radiation (IR) could be combined with systemically delivered oncolytic vaccinia virus. EXPERIMENTAL DESIGN: U-87 glioma xenografts were grown subcutaneously or orthotopically. Oncolytic vaccinia viruses GLV-1h68 and LIVP 1.1.1 were injected systemically and IR was given focally to glioma xenografts. In a bilateral tumor model, glioma xenografts were grown in both flanks, oncolytic vaccinia was injected systemically and radiation was delivered specifically to the right flank tumor, whereas the left flank tumor was shielded. Viral replication and tumor regression, after systemic injection, was analyzed and compared in irradiated and nonirradiated glioma xenografts. RESULTS: Systemically administered oncolytic vaccinia virus replicated to higher titers in preirradiated U-87 xenografts than in nonirradiated glioma xenografts. This increased oncolytic viral replication correlated with increased tumor xenograft regression and mouse survival in subcutaneous and orthotopic U-87 glioma models compared with monotherapies. The ability of focal IR to mediate selective replication of oncolytic vaccinia was shown in a bilateral glioma model in which systemically administered oncolytic vaccinia replicated preferentially in the irradiated tumor compared with the nonirradiated tumor in the same mouse. CONCLUSION: These findings show a potential clinical role of focal IR in sensitizing irradiated tumor sites for preferential vaccinia virus-mediated oncolysis.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Viroterapia Oncolítica , Radiación Ionizante , Vaccinia/terapia , Replicación Viral , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Células Cultivadas , Chlorocebus aethiops , Terapia Combinada , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Glioma/genética , Glioma/mortalidad , Riñón/citología , Riñón/metabolismo , Riñón/efectos de la radiación , Masculino , Ratones , Ratones Desnudos , Tasa de Supervivencia , Vaccinia/genética , Vaccinia/mortalidad , Virus Vaccinia/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Surg Res ; 175(2): e53-60, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22341347

RESUMEN

BACKGROUND: Approximately one fourth of bone and soft-tissue sarcomas recur after prior treatment. GLV-1h68 is a recombinant, replication-competent vaccinia virus that has been shown to have oncolytic effects against many human cancer types. We sought to determine whether GLV-1h68 could selectively target and lyse a panel of human bone and soft-tissue sarcoma cell lines in vitro and in vivo. METHODS: GLV-1h68 was tested in a panel of four cell lines including: fibrosarcoma HT-1080, osteosarcoma U-2OS, fibrohistiocytoma M-805, and rhabdomyosarcoma HTB-82. Gene expression, infectivity, viral proliferation, and cytotoxicity were characterized in vitro. HT-1080 xenograft flank tumors grown in vivo were injected intratumorally with a single dose of GLV-1h68. RESULTS: All four cell lines supported robust viral transgene expression in vitro. At a multiplicity of infection (MOI) of five, GLV-1h68 was cytotoxic to three cell lines, resulting in >80% cytotoxicity over 7 d. In vivo, a single injection of GLV-1h68 into HT-1080 xenografts exhibited localized intratumoral luciferase activity peaking at d 2-4, with gradual resolution over 8 d and no evidence of spread to normal tissues. Treated animals exhibited near-complete tumor regression over a 28-d period without observed toxicity. CONCLUSION: GLV-1h68 has potent direct oncolytic effects against human sarcoma in vitro and in vivo. Recombinant vaccinia oncolytic virotherapy could provide a new platform for the treatment of patients with bone and soft tissue sarcomas. Future clinical trials investigating oncolytic vaccinia as a therapy for sarcomas are warranted.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Vacunas contra el Cáncer/uso terapéutico , Viroterapia Oncolítica , Sarcoma/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Virus Vaccinia , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Histiocitoma Fibroso Maligno/tratamiento farmacológico , Histiocitoma Fibroso Maligno/patología , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Desnudos , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Sarcoma/patología , Neoplasias de los Tejidos Blandos/patología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Transl Med ; 10: 9, 2012 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-22236378

RESUMEN

BACKGROUND: Combination of oncolytic vaccinia virus therapy with conventional chemotherapy has shown promise for tumor therapy. However, side effects of chemotherapy including thrombocytopenia, still remain problematic. METHODS: Here, we describe a novel approach to optimize combination therapy of oncolytic virus and chemotherapy utilizing virus-encoding hyper-IL-6, GLV-1h90, to reduce chemotherapy-associated side effects. RESULTS: We showed that the hyper-IL-6 cytokine was successfully produced by GLV-1h90 and was functional both in cell culture as well as in tumor-bearing animals, in which the cytokine-producing vaccinia virus strain was well tolerated. When combined with the chemotherapeutic mitomycin C, the anti-tumor effect of the oncolytic virotherapy was significantly enhanced. Moreover, hyper-IL-6 expression greatly reduced the time interval during which the mice suffered from chemotherapy-induced thrombocytopenia. CONCLUSION: Therefore, future clinical application would benefit from careful investigation of additional cytokine treatment to reduce chemotherapy-induced side effects.


Asunto(s)
Plaquetas/efectos de los fármacos , Interleucina-6/farmacología , Mitomicina/toxicidad , Neoplasias/terapia , Neoplasias/virología , Viroterapia Oncolítica/efectos adversos , Virus Vaccinia/fisiología , Animales , Línea Celular Tumoral , Terapia Combinada , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Inyecciones , Interleucina-6/sangre , Quinasas Janus/metabolismo , Masculino , Ratones , Ratones Desnudos , Mitomicina/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Virus Vaccinia/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Ann Surg Oncol ; 19 Suppl 3: S665-74, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22258815

RESUMEN

BACKGROUND: Replication-competent oncolytic viruses have shown great promise as a potential cancer treatment. This study aimed to determine whether a novel vaccinia virus, GLV-1h151, with genetic modifications enhancing cancer specificity and enabling virus detection, is effective against a range of human cancers and is safe when administered in preclinical models. METHODS: GLV-1h151 was modified with deletion of thymidine kinase enhancing specificity and insertion of the green fluorescent protein (GFP) gene. The virus was tested in several human cancer cell lines for cytotoxicity including breast, lung, pancreatic, and colorectal. Virus replication was assessed via visualization of GFP expression and bioluminescence, and viral plaque assays. Finally, GLV-1h151 was administered systemically or intratumorally in mice with pancreatic cancer xenografts (PANC-1) to assess virus biodistribution, toxicity, and effect on tumor growth. RESULTS: GLV-1h151 effectively infected, replicated in, and killed several cancer cell types. Detection and visualization of virus replication was successful via fluorescence imaging of GFP expression, which was dose dependent. When administered intravenously or intratumorally in vivo, GLV-1h151 regressed tumor growth (P < 0.001) and displayed a good biosafety profile. GLV-1h151 infection and replication in tumors was successfully visualized via GFP and bioluminescence, with virus presence in tumors confirmed histologically. CONCLUSIONS: GLV-1h151 is effective as an oncolytic agent against a wide range of cancers in cell culture and is effective against pancreatic human xenografts displaying a good biosafety profile and ability to be detected via optical imaging. GLV-1h151 thus adds another potential medium for the killing of cancer and detection of virus in infected tissue.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Neoplasias Pancreáticas/terapia , Virus Vaccinia/fisiología , Animales , Línea Celular Tumoral , Supervivencia Celular , Citometría de Flujo , Ingeniería Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inyecciones Intralesiones , Inyecciones Intravenosas , Masculino , Ratones , Ratones Desnudos , Neoplasias/terapia , Virus Oncolíticos/genética , Neoplasias Pancreáticas/virología , Virus Vaccinia/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Transl Med ; 9: 164, 2011 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-21951588

RESUMEN

BACKGROUND: We have shown that insertion of the three vaccinia virus (VACV) promoter-driven foreign gene expression cassettes encoding Renilla luciferase-Aequorea GFP fusion protein, ß-galactosidase, and ß-glucuronidase into the F14.5L, J2R, and A56R loci of the VACV LIVP genome, respectively, results in a highly attenuated mutant strain GLV-1h68. This strain shows tumor-specific replication and is capable of eradicating tumors with little or no virulence in mice. This study aimed to distinguish the contribution of added VACV promoter-driven transcriptional units as inserts from the effects of insertional inactivation of three viral genes, and to determine the correlation between replication efficiency of oncolytic vaccinia virus in cell cultures and the virulence and antitumor efficacy in mice METHODS: A series of recombinant VACV strains was generated by replacing one, two, or all three of the expression cassettes in GLV-1h68 with short non-coding DNA sequences. The replication efficiency and tumor cell killing capacity of these newly generated VACV strains were compared with those of the parent virus GLV-1h68 in cell cultures. The virus replication efficiency in tumors and antitumor efficacy as well as the virulence were evaluated in nu/nu (nude) mice bearing human breast tumor xenografts. RESULTS: we found that virus replication efficiency increased with removal of each of the expression cassettes. The increase in virus replication efficiency was proportionate to the strength of removed VACV promoters linked to foreign genes. The replication efficiency of the new VACV strains paralleled their cytotoxicity in cell cultures. The increased replication efficiency in tumor xenografts resulted in enhanced antitumor efficacy in nude mice. Similarly, the enhanced virus replication efficiency was indicative of increased virulence in nude mice. CONCLUSIONS: These data demonstrated that insertion of VACV promoter-driven transcriptional units into the viral genome for the purpose of insertional mutagenesis did modulate the efficiency of virus replication together with antitumor efficacy as well as virulence. Replication efficiency of oncolytic VACV in cell cultures can predict the virulence and therapeutic efficacy in nude mice. These findings may be essential for rational design of safe and potent VACV strains for vaccination and virotherapy of cancer in humans and animals.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Virus Oncolíticos/patogenicidad , Virus Vaccinia/fisiología , Virus Vaccinia/patogenicidad , Replicación Viral/fisiología , Animales , Secuencia de Bases , Técnicas de Cultivo de Célula , Muerte Celular , Línea Celular Tumoral , ADN Intergénico/genética , Expresión Génica , Genoma Viral , Humanos , Ratones , Ratones Desnudos , Mutagénesis Insercional/genética , Virus Oncolíticos/genética , Regiones Promotoras Genéticas/genética , Resultado del Tratamiento , Virus Vaccinia/genética , Virulencia , Ensayos Antitumor por Modelo de Xenoinjerto
19.
PLoS One ; 6(7): e22069, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21779374

RESUMEN

Virotherapy using oncolytic vaccinia virus strains is one of the most promising new strategies for cancer therapy. In this study, we analyzed for the first time the therapeutic efficacy of the oncolytic vaccinia virus GLV-1h68 in two human hepatocellular carcinoma cell lines HuH7 and PLC/PRF/5 (PLC) in cell culture and in tumor xenograft models. By viral proliferation assays and cell survival tests, we demonstrated that GLV-1h68 efficiently colonized, replicated in, and did lyse these cancer cells in culture. Experiments with HuH7 and PLC xenografts have revealed that a single intravenous injection (i.v.) of mice with GLV-1h68 resulted in a significant reduction of primary tumor sizes compared to uninjected controls. In addition, replication of GLV-1h68 in tumor cells led to strong inflammatory and oncolytic effects resulting in intense infiltration of MHC class II-positive cells like neutrophils, macrophages, B cells and dendritic cells and in up-regulation of 13 pro-inflammatory cytokines. Furthermore, GLV-1h68 infection of PLC tumors inhibited the formation of hemorrhagic structures which occur naturally in PLC tumors. Interestingly, we found a strongly reduced vascular density in infected PLC tumors only, but not in the non-hemorrhagic HuH7 tumor model. These data demonstrate that the GLV-1h68 vaccinia virus may have an enormous potential for treatment of human hepatocellular carcinoma in man.


Asunto(s)
Carcinoma Hepatocelular/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Virus Vaccinia/fisiología , Animales , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Chlorocebus aethiops , Citometría de Flujo , Humanos , Neoplasias Hepáticas/terapia , Ratones , Ratones Desnudos , Virus Oncolíticos/genética , Virus Vaccinia/genética , Replicación Viral
20.
J Transl Med ; 9: 36, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21453532

RESUMEN

INTRODUCTION: Oncolytic viruses show promise for treating cancer. However, to assess therapeutic efficacy and potential toxicity, a noninvasive imaging modality is needed. This study aimed to determine if insertion of the human sodium iodide symporter (hNIS) cDNA as a marker for non-invasive imaging of virotherapy alters the replication and oncolytic capability of a novel vaccinia virus, GLV-1h153. METHODS: GLV-1h153 was modified from parental vaccinia virus GLV-1h68 to carry hNIS via homologous recombination. GLV-1h153 was tested against human pancreatic cancer cell line PANC-1 for replication via viral plaque assays and flow cytometry. Expression and transportation of hNIS in infected cells was evaluated using Westernblot and immunofluorescence. Intracellular uptake of radioiodide was assessed using radiouptake assays. Viral cytotoxicity and tumor regression of treated PANC-1tumor xenografts in nude mice was also determined. Finally, tumor radiouptake in xenografts was assessed via positron emission tomography (PET) utilizing carrier-free 124I radiotracer. RESULTS: GLV-1h153 infected, replicated within, and killed PANC-1 cells as efficiently as GLV-1h68. GLV-1h153 provided dose-dependent levels of hNIS expression in infected cells. Immunofluorescence detected transport of the protein to the cell membrane prior to cell lysis, enhancing hNIS-specific radiouptake (P < 0.001). In vivo, GLV-1h153 was as safe and effective as GLV-1h68 in regressing pancreatic cancer xenografts (P < 0.001). Finally, intratumoral injection of GLV-1h153 facilitated imaging of virus replication in tumors via 124I-PET. CONCLUSION: Insertion of the hNIS gene does not hinder replication or oncolytic capability of GLV-1h153, rendering this novel virus a promising new candidate for the noninvasive imaging and tracking of oncolytic viral therapy.


Asunto(s)
Mutagénesis Insercional/genética , Virus Oncolíticos/fisiología , Tomografía de Emisión de Positrones , Simportadores/genética , Virus Vaccinia/fisiología , Replicación Viral/fisiología , Animales , Western Blotting , Muerte Celular , Línea Celular , Membrana Celular/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Radioisótopos de Yodo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Simportadores/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...