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










Base de dados
Intervalo de ano de publicação
1.
Head Neck ; 38(2): 237-46, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25244076

RESUMO

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.


Assuntos
Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Neoplasias da Glândula Tireoide/imunologia , Vaccinia virus , Animais , Biopolímeros , Linhagem Celular Tumoral , Fibroínas , Fibronectinas , Cuidados Intraoperatórios , Camundongos Nus , Proteínas Recombinantes de Fusão , Neoplasias da Glândula Tireoide/cirurgia , Ensaio de Placa Viral , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Galactosidase/metabolismo
2.
Surgery ; 158(2): 331-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26049609

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/terapia , Terapia Genética , Neoplasias Hepáticas/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Vaccinia virus/genética , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
3.
PLoS One ; 9(8): e104337, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25093734

RESUMO

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.


Assuntos
Vetores Genéticos/genética , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Vírus Oncolíticos/genética , Vaccinia virus/genética , Animais , Gatos , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Chlorocebus aethiops , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/terapia , Camundongos , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Terapia Viral Oncolítica , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Surgery ; 156(2): 263-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24957667

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Niacinamida/análogos & derivados , Terapia Viral Oncolítica/métodos , Compostos de Fenilureia/farmacologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Engenharia Genética , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Niacinamida/farmacologia , Sorafenibe , Vaccinia virus/fisiologia , Ensaio de Placa Viral , Replicação Viral
5.
PLoS One ; 8(9): e71105, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24019862

RESUMO

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.


Assuntos
Líquidos Corporais , Metástase Neoplásica/terapia , Terapia Viral Oncolítica , Vaccinia virus/fisiologia , Animais , Sequência de Bases , Primers do DNA , Xenoenxertos , Humanos , Masculino , Camundongos , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia
6.
Int J Cancer ; 133(12): 2989-99, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23729266

RESUMO

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.


Assuntos
Endotélio Vascular/efeitos da radiação , Glioma/terapia , Terapia Viral Oncolítica/métodos , Tolerância a Radiação , Vaccinia virus/genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Glioma/irrigação sanguínea , Humanos , Masculino , Camundongos , Fator A de Crescimento do Endotélio Vascular/fisiologia
7.
Mol Ther ; 21(11): 2054-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23765443

RESUMO

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.


Assuntos
Anemia/terapia , Eritropoetina/metabolismo , Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vaccinia virus/genética , Anemia/complicações , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Eritropoetina/genética , Proteínas de Fluorescência Verde , Humanos , Neoplasias Hepáticas Experimentais , Masculino , Camundongos , Camundongos Nus , Microvasos/metabolismo , Vírus Oncolíticos/metabolismo , Proteínas Recombinantes/metabolismo , Vaccinia virus/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
JAMA Otolaryngol Head Neck Surg ; 139(2): 173-82, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23429949

RESUMO

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.


Assuntos
Adenocarcinoma/terapia , Carcinoma Mucoepidermoide/terapia , Terapia Viral Oncolítica , Neoplasias das Glândulas Salivares/terapia , Vaccinia virus/fisiologia , Adenocarcinoma/patologia , Animais , Carcinoma Mucoepidermoide/patologia , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Injeções , Luciferases/genética , Camundongos , Camundongos Nus , Neoplasias das Glândulas Salivares/patologia , Vaccinia virus/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Galactosidase/genética
9.
Bioengineered ; 4(2): 84-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23093804

RESUMO

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.


Assuntos
Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Vaccinia virus/fisiologia , Animais , Cães , Camundongos , Neoplasias/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vaccinia virus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Nucl Med ; 53(12): 1933-42, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23139088

RESUMO

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.


Assuntos
Imagem Molecular/métodos , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Radioterapia Guiada por Imagem/métodos , Simportadores/genética , Vaccinia virus/genética , Animais , Linhagem Celular Tumoral , Estudos de Viabilidade , Humanos , Radioisótopos do Iodo , Camundongos , Imagem Óptica , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/radioterapia , Tomografia por Emissão de Pósitrons , Fatores de Tempo , Resultado do Tratamento , Carga Tumoral
11.
Hum Gene Ther Methods ; 23(5): 285-96, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23131031

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Vírus Oncolíticos/fisiologia , Transcrição Gênica , Vaccinia virus/fisiologia , Replicação Viral , Linhagem Celular , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação Viral da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Transdução de Sinais
12.
Methods Mol Biol ; 872: 159-75, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22700410

RESUMO

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.


Assuntos
Diagnóstico por Imagem/métodos , Proteínas Luminescentes/metabolismo , Animais , Bactérias/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia de Fluorescência , Neoplasias , Ratos , Vaccinia virus/metabolismo
13.
PLoS One ; 7(5): e37239, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615950

RESUMO

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.


Assuntos
Neoplasias Musculares/veterinária , Terapia Viral Oncolítica/métodos , Sarcoma/veterinária , Neoplasias de Tecidos Moles/veterinária , Vaccinia virus/fisiologia , Animais , Neoplasias Ósseas/secundário , Neoplasias Ósseas/veterinária , Linhagem Celular Tumoral , Cães , Camundongos , Neoplasias Musculares/terapia , Vírus Oncolíticos/fisiologia , Sarcoma/patologia , Sarcoma/terapia , Neoplasias de Tecidos Moles/terapia , Replicação Viral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Clin Cancer Res ; 18(9): 2579-90, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22379115

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Terapia Viral Oncolítica , Radiação Ionizante , Vacínia/terapia , Replicação Viral , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Células Cultivadas , Chlorocebus aethiops , Terapia Combinada , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Glioma/genética , Glioma/mortalidade , Rim/citologia , Rim/metabolismo , Rim/efeitos da radiação , Masculino , Camundongos , Camundongos Nus , Taxa de Sobrevida , Vacínia/genética , Vacínia/mortalidade , Vaccinia virus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Surg Res ; 175(2): e53-60, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22341347

RESUMO

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.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Vacinas Anticâncer/uso terapêutico , Terapia Viral Oncolítica , Sarcoma/tratamento farmacológico , Neoplasias de Tecidos Moles/tratamento farmacológico , Vaccinia virus , Animais , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/patologia , Histiocitoma Fibroso Maligno/tratamento farmacológico , Histiocitoma Fibroso Maligno/patologia , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Nus , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Rabdomiossarcoma/tratamento farmacológico , Rabdomiossarcoma/patologia , Sarcoma/patologia , Neoplasias de Tecidos Moles/patologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Ann Surg Oncol ; 19 Suppl 3: S665-74, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22258815

RESUMO

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.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Neoplasias Pancreáticas/terapia , Vaccinia virus/fisiologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Citometria de Fluxo , Engenharia Genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Injeções Intralesionais , Injeções Intravenosas , Masculino , Camundongos , Camundongos Nus , Neoplasias/terapia , Vírus Oncolíticos/genética , Neoplasias Pancreáticas/virologia , Vaccinia virus/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Transl Med ; 10: 9, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-22236378

RESUMO

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.


Assuntos
Plaquetas/efeitos dos fármacos , Interleucina-6/farmacologia , Mitomicina/toxicidade , Neoplasias/terapia , Neoplasias/virologia , Terapia Viral Oncolítica/efeitos adversos , Vaccinia virus/fisiologia , Animais , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Injeções , Interleucina-6/sangue , Janus Quinases/metabolismo , Masculino , Camundongos , Camundongos Nus , Mitomicina/uso terapêutico , Neoplasias/tratamento farmacológico , Proteínas Recombinantes/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vaccinia virus/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Transl Med ; 9: 164, 2011 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-21951588

RESUMO

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.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Vírus Oncolíticos/patogenicidade , Vaccinia virus/fisiologia , Vaccinia virus/patogenicidade , Replicação Viral/fisiologia , Animais , Sequência de Bases , Técnicas de Cultura de Células , Morte Celular , Linhagem Celular Tumoral , DNA Intergênico/genética , Expressão Gênica , Genoma Viral , Humanos , Camundongos , Camundongos Nus , Mutagênese Insercional/genética , Vírus Oncolíticos/genética , Regiões Promotoras Genéticas/genética , Resultado do Tratamento , Vaccinia virus/genética , Virulência , Ensaios Antitumorais Modelo de Xenoenxerto
19.
PLoS One ; 6(7): e22069, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21779374

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Vaccinia virus/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Chlorocebus aethiops , Citometria de Fluxo , Humanos , Neoplasias Hepáticas/terapia , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Vaccinia virus/genética , Replicação Viral
20.
J Transl Med ; 9: 36, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21453532

RESUMO

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.


Assuntos
Mutagênese Insercional/genética , Vírus Oncolíticos/fisiologia , Tomografia por Emissão de Pósitrons , Simportadores/genética , Vaccinia virus/fisiologia , Replicação Viral/fisiologia , Animais , Western Blotting , Morte Celular , Linhagem Celular , Membrana Celular/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Radioisótopos do Iodo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Simportadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...