Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Ther ; 20(6): 1139-47, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22334023

RESUMO

Helicobacter pylori neutrophil-activating protein (NAP) is a major virulence factor and powerful inducer of inflammatory reaction and Th1-polarized immune response. Here, we evaluated the therapeutic efficacy of measles virus (MV) strains engineered to express secretory NAP forms against metastatic breast cancer. Recombinant viruses encoding secretory NAP forms (MV-lambda-NAP and MV-s-NAP) efficiently infect and destroy breast cancer cells by cell-to-cell viral spread and large syncytia formation independently of hormone receptor status. Intrapleural administration of MV-s-NAP doubled the median survival in a pleural effusion xenograft model: 65 days as compared to 29 days in the control group (P < 0.0001). This therapeutic effect correlated with a brisk Th1 type cytokine response in vivo. Secretory NAP was expressed at high levels by infected tumor cells and increased tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-12/23 cytokine concentrations were detected in the pleural effusion. In an aggressive model of lung metastatic breast cancer, MV-lambda-NAP and MV-s-NAP also significantly improved survival of the treated animals (P < 0.05) as compared to the control MV strain. These data suggest that potent immunomodulators of bacterial origin, such as H. pylori NAP, can enhance the antitumor effect of oncolytic viruses and support the feasibility and potential of a combined viroimmunotherapy approach.


Assuntos
Proteínas de Bactérias/genética , Neoplasias da Mama/terapia , Helicobacter pylori , Fatores Imunológicos/genética , Vírus do Sarampo/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Ordem dos Genes , Helicobacter pylori/imunologia , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Nus , Camundongos Transgênicos , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Derrame Pleural Maligno/imunologia , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cancer Gene Ther ; 12(7): 593-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15746945

RESUMO

Oncolytic viruses are promising cytoreductive agents for cancer treatment but extensive human testing will be required before they are made commercially available. Here, we investigated the oncolytic potential of two commercially available live attenuated vaccines, Moraten measles and Jeryl-Lynn mumps, in a murine model of intraperitoneal human ovarian cancer and compared their efficacies against a recombinant oncolytic measles virus (MV-CEA) that is being tested in a phase I clinical trial. The common feature of these viruses is that they express hemagglutinin and fusion therapeutic proteins that can induce extensive fusion of the infected cell with its neighbors, resulting in death of the cell monolayer. In vitro, the three viruses caused intercellular fusion in human ovarian cancer cells but with marked differences in fusion kinetics. MV-CEA was the fastest followed by Jeryl-Lynn mumps virus while Moraten measles virus was the slowest, although all viruses eventually caused comparable cell death 6 days postinfection. Tumor-bearing mice treated with 10(6) or 10(7) pfu (one thousand times the vaccine dose) of each of the three viruses responded favorably to therapy with significant prolongations in survival. All three viruses demonstrated equivalent antitumor potency. Commercially available Moraten measles and Jeryl-Lynn mumps vaccines warrant further investigation as potential anticancer agents.


Assuntos
Vacina contra Sarampo/uso terapêutico , Vacina contra Caxumba/uso terapêutico , Neoplasias Ovarianas/terapia , Animais , Antígeno Carcinoembrionário/sangue , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Efeito Citopatogênico Viral , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Células Tumorais Cultivadas , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Hum Gene Ther ; 14(16): 1565-77, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14577918

RESUMO

In support of a proposed phase I clinical trial, we studied the biodistribution of virus-infected cells after intraperitoneal administration of oncolytic measles viruses to alpha/beta interferon-defective mice expressing human CD46 with human-like tissue specificity. Various marker genes were employed, and green fluorescent protein proved to be most informative. Mesothelium and ovarian surface epithelium were remarkably resistant to infection, but infected peritoneal macrophages were present in abundance both in peritoneal lavage fluid and in the greater omentum, where they were heavily concentrated in "milky spots". Infected macrophages were also identified outside the peritoneal cavity, along the peritoneal fluid drainage pathway and in the spleen. Thus, diaphragmatic stomata, thoracic lymphatic vessels, and parathymic lymph nodes contained numerous measles-infected cells, as did the marginal zones of the white pulp of the spleen. Splenic marginal zone macrophages were the predominant targets of infection after intravenous administration of oncolytic measles viruses. When measles-infected peritoneal macrophages were adoptively transferred, they did not migrate beyond the confines of the peritoneal cavity, suggesting that, after intraperitoneal virus administration, the positive cells in thoracic lymphatics, parathymic lymph nodes, and spleen are nonmigratory cells transduced in situ by viral particles that have exited from the peritoneal cavity.


Assuntos
Antígenos CD/fisiologia , Injeções Intraperitoneais , Vírus do Sarampo/fisiologia , Glicoproteínas de Membrana/fisiologia , Cavidade Peritoneal/citologia , Proteínas Recombinantes/administração & dosagem , Animais , Proteínas de Fluorescência Verde , Interferon Tipo I/metabolismo , Proteínas Luminescentes/metabolismo , Macrófagos/virologia , Proteína Cofatora de Membrana , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Cavidade Peritoneal/virologia
4.
Head Neck ; 36(11): 1619-27, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24115092

RESUMO

BACKGROUND: Recombinant vesicular stomatitis virus expressing interferon-ß (VSV-IFN-ß) has demonstrated antitumor activity in vitro and in vivo. In preparation for clinical testing in human squamous cell carcinoma (SCC) of the head and neck, we conducted preclinical studies of VSV-IFN-ß in syngeneic SCC models. METHODS: In vitro, VSV-IFN-ß (expressing rat or mouse interferon [IFN]-ß)-induced cytotoxicity and propagated in rat (FAT-7) or mouse (SCC-VII) SCC cells during normoxia and hypoxia. In vivo, intratumoral administration of VSV-rat-IFN-ß or VSV-human-IFN-ß in FAT-7 bearing or non-tumor bearing immunocompetent rats did not result in acute organ toxicity or death. RESULTS: VSV-r-IFN-ß replicated predominantly in tumors and a dose dependent anti-VSV antibody response was observed. Intratumoral or intravenous administration of VSV-IFN-ß resulted in growth delay and improved survival compared with controls. CONCLUSION: The above data confirm safety and feasibility of VSV-IFN-ß administration in immunocompetent animals and support its clinical evaluation in advanced human head and neck cancer.


Assuntos
Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/terapia , Interferon beta/farmacologia , Terapia Viral Oncolítica/métodos , Vesiculovirus/isolamento & purificação , Análise de Variância , Animais , Western Blotting , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Estudos de Viabilidade , Feminino , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imunocompetência , Estimativa de Kaplan-Meier , Camundongos , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Segurança , Estatísticas não Paramétricas , Transplante Isogênico , Resultado do Tratamento , Células Tumorais Cultivadas , Estomatite Vesicular/virologia
5.
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
6.
Expert Opin Biol Ther ; 8(2): 213-20, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18194077

RESUMO

BACKGROUND: Recurrent gliomas have a dismal outcome despite use of multimodality treatment including surgery, radiation therapy and chemotherapy. OBJECTIVE: In this article the authors discuss potential applications of oncolytic measles virus strains as novel antitumor agents in the treatment of gliomas. METHODS: Important aspects of measles virus development as an anticancer therapeutic agent including engineering, retargeting and combination studies with other therapeutic modalities are discussed. The translational process that led to the first clinical trial of an engineered measles virus derivative in patients with recurrent glioblastoma multiforme is also described. RESULTS/CONCLUSIONS: Oncolytic measles virus strains hold promise as novel antitumor agents in the treatment of gliomas.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Vacina contra Sarampo/uso terapêutico , Vírus do Sarampo/imunologia , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Glioma/genética , Glioma/imunologia , Humanos , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Vírus do Sarampo/genética , Vírus Oncolíticos/genética
7.
Hum Gene Ther ; 19(7): 690-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18576918

RESUMO

Gliomas have a dismal prognosis, with the median survival of patients with the most common histology, glioblastoma multiforme, being only 12-15 months. Development of novel therapeutic agents is urgently needed. We have previously demonstrated that oncolytic measles virus strains derived from the Edmonston vaccine lineage have significant antitumor activity against gliomas [Phuong, L.K., Allen, C., Peng, K.W., Giannini, C., Greiner, S., Teneyck, C.J., Mishra, P.K., Macura, S.I., Russell, S.J., Galanis, E.C. (2003). Cancer. Res. 63, 2462-2469]. MV-CEA is an Edmonston vaccine lineage measles virus strain engineered to express the marker peptide carcinoembryonic antigen (CEA): CEA levels can serve as a correlate of viral gene expression. In support of a phase I clinical trial of intratumoral and resection cavity administration of MV-CEA to patients with recurrent gliomas, we assessed the neurotoxicity of MV-CEA in adult immune male rhesus macaques (Macaca mulatta). The animals ' immune status and administration schedule mimicked the trial population and proposed administration schema. Macaca mulatta represents the prototype animal species for assessment of measles neurotoxicity. The animals were stereotactically administered either vehicle (n = 1) or MV-CEA at 2 x 10(5)or 2 x 10(6) TCID(50) (each, n = 2) in the right frontal lobe in two injections on days 1 and 5. Macaques were closely monitored clinically for neurotoxicity. Body weight, temperature, complete blood count, CEA, clinical chemistries, coagulation, complement levels, immunoglobulin, measles antibody titers, viremia, and shedding (buccal swabs) were tested at multiple time points. Furthermore, cisterna magna spinal taps were performed on day 9 and 1 year after the first viral dose administration, and samples were analyzed for protein, glucose, cell differential, and presence of MV-CEA. Magnetic resonance imaging (MRI) was performed between 4 and 5 months after article administration to assess for subclinical neurotoxicity. To date, 36+ months from study initiation there has been no clinical or biochemical evidence of toxicity, including lack of neurological symptoms, fever, or other systemic symptoms and lack of immunosuppression. Quantitative RT-PCR analysis of blood, buccal swabs, and cerebrospinal fluid (CSF) was negative for MV-CEA at all time points, with the exception of viral genome deletion in the blood of one asymptomatic animal at the 2 x 10(6) TCID(50) dose level on day 85. Vero cell overlays of CSF cells and supernatant were negative for viral recovery. There was no detection of CEA in serum or CSF at any time point. MRI scans were negative for imaging abnormalities and showed no evidence of encephalitis. Our results support the safety of CNS administration of MV-CEA in glioma patients. A clinical trial of intratumoral and resection cavity administration of MV-CEA in patients with recurrent glioblastoma multiforme is currently ongoing.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer , Antígeno Carcinoembrionário/metabolismo , Vetores Genéticos , Glioma/terapia , Vírus do Sarampo , Animais , Encéfalo/virologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/prevenção & controle , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/genética , Chlorocebus aethiops , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Efeito Citopatogênico Viral , Vias de Administração de Medicamentos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Glioma/genética , Glioma/patologia , Glioma/prevenção & controle , Humanos , Macaca mulatta , Masculino , Vírus do Sarampo/genética , Vírus do Sarampo/patogenicidade , Recidiva , Resultado do Tratamento , Células Vero
8.
Hepatology ; 44(6): 1465-77, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17133484

RESUMO

The oncolytic measles virus Edmonston strain (MV-Edm), a nonpathogenic virus targeting cells expressing abundant CD46, selectively destroys neoplastic tissue. Clinical development of MV-Edm would benefit from noninvasive monitoring strategies to determine the speed and extent of the spread of the virus in treated patients and the location of virus-infected cells. We evaluated recombinant MV-Edm expressing carcinoembryonic antigen (CEA) or the human sodium iodide symporter (hNIS) for oncolytic potential in hepatocellular carcinoma (HCC) and efficiency in tracking viruses in vivo by noninvasive monitoring. CD46 expression in human HCC and primary hepatocytes was assessed by flow cytometry and immunohistochemistry. Infectivity, syncytium formation, and cytotoxicity of recombinant MV-Edm in HCC cell lines were evaluated by fluorescence microscopy, crystal violet staining, and the MTS assay. Transgene expression in HCC cell lines after infection with recombinant MV-Edm in vitro and in vivo was assessed by CEA concentration, 125I-uptake, and 123I-imaging studies. Toxicology studies were performed in Ifnar(KO)xCD46 transgenic mice. The CD46 receptor was highly expressed in HCC compared to nonmalignant hepatic tissue. Recombinant MV-Edm efficiently infected HCC cell lines, resulting in extensive syncytium formation followed by cell death. Transduction of HCC cell lines and subcutaneous HCC xenografts with recombinant MV-Edm resulted in high-level expression of transgenes in vitro and in vivo. MV-Edm was nontoxic in susceptible mice. Intratumoral and intravenous therapy with recombinant MV-Edm resulted in inhibition of tumor growth and prolongation of survival with complete tumor regression in up to one third of animals. In conclusion, engineered MV-Edm may be a potent and novel cancer gene therapy system for HCC. MV-Edm expressing CEA or hNIS elicited oncolytic effects in human HCC cell lines in vitro and in vivo, enabling the spread of the virus to be monitored in a noninvasive manner.


Assuntos
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Vírus do Sarampo/genética , Terapia Viral Oncolítica/métodos , Animais , Antígeno Carcinoembrionário/genética , Efeito Citopatogênico Viral , Feminino , Engenharia Genética/métodos , Humanos , Injeções Intravenosas , Radioisótopos do Iodo/metabolismo , Masculino , Vacina contra Sarampo/genética , Vírus do Sarampo/imunologia , Proteína Cofatora de Membrana/biossíntese , Camundongos , Transplante de Neoplasias , Simportadores/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Mol Ther ; 12(5): 835-41, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16054438

RESUMO

The sodium-iodide symporter (NIS) is primarily a thyroid protein, providing for the accumulation of iodide for biosynthesis of thyroid hormones. Native NIS expression has made possible the use of radioactive iodide to image and treat thyroid disease successfully. The current study, using adult male beagle dogs, was carried out in preparation for a Phase I clinical trial of adenovirus-mediated NIS gene (approved symbol SLC5A5) therapy for prostate cancer. Direct intraprostatic injection of virus (Ad5/CMV/NS) was followed by iv injection of 3 mCi 123I and serial image acquisition. The dogs were then given a therapeutic dose of 131I (116 mCi/m2) and observed for 7 days. SPECT/CT fusion imaging revealed clear images of the NIS-transduced prostates. Dosimetry calculations revealed an average absorbed dose to the prostate of 23 +/- 42 cGy/mCi 131I, with acceptably low radiation doses to other organs. This study demonstrated the successful introduction of localized NIS expression in the prostate gland of dogs, with no vector-related toxicity observed. None of the animals experienced any surgical complications, and serum chemistry panels showed no significant change following therapy. The results presented provide further evidence of the safety and efficacy of NIS as a therapeutic gene and support translation of this work into the clinical setting.


Assuntos
Adenoviridae/genética , Terapia Genética , Neoplasias da Próstata/terapia , Simportadores/uso terapêutico , Animais , Modelos Animais de Doenças , Cães , Radioisótopos do Iodo/uso terapêutico , Masculino , Neoplasias da Próstata/diagnóstico por imagem , Radiometria , Cintilografia , Compostos Radiofarmacêuticos/uso terapêutico , Simportadores/genética , Transdução Genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA