Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Commun ; 15(1): 493, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216554

RESUMO

Measles virus (MV) vaccine strains have shown significant preclinical antitumor activity against glioblastoma (GBM), the most lethal glioma histology. In this first in human trial (NCT00390299), a carcinoembryonic antigen-expressing oncolytic measles virus derivative (MV-CEA), was administered in recurrent GBM patients either at the resection cavity (Group A), or, intratumorally on day 1, followed by a second dose administered in the resection cavity after tumor resection on day 5 (Group B). A total of 22 patients received study treatment, 9 in Group A and 13 in Group B. Primary endpoint was safety and toxicity: treatment was well tolerated with no dose-limiting toxicity being observed up to the maximum feasible dose (2×107 TCID50). Median OS, a secondary endpoint, was 11.6 mo and one year survival was 45.5% comparing favorably with contemporary controls. Other secondary endpoints included assessment of viremia, MV replication and shedding, humoral and cellular immune response to the injected virus. A 22 interferon stimulated gene (ISG) diagonal linear discriminate analysis (DLDA) classification algorithm in a post-hoc analysis was found to be inversely (R = -0.6, p = 0.04) correlated with viral replication and tumor microenvironment remodeling including proinflammatory changes and CD8 + T cell infiltration in post treatment samples. This data supports that oncolytic MV derivatives warrant further clinical investigation and that an ISG-based DLDA algorithm can provide the basis for treatment personalization.


Assuntos
Glioblastoma , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Vírus do Sarampo/genética , Antígeno Carcinoembrionário/genética , Recidiva Local de Neoplasia/terapia , Vacina contra Sarampo , Microambiente Tumoral
2.
Mol Ther Methods Clin Dev ; 26: 532-546, 2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36092362

RESUMO

Despite recent therapeutic advances, metastatic breast cancer (MBC) remains incurable. Engineered measles virus (MV) constructs based on the attenuated MV Edmonston vaccine platform have demonstrated significant oncolytic activity against solid tumors. The Helicobacter pylori neutrophil-activating protein (NAP) is responsible for the robust inflammatory reaction in gastroduodenal mucosa during bacterial infection. NAP attracts and activates immune cells at the site of infection, inducing expression of pro-inflammatory mediators. We engineered an MV strain to express the secretory form of NAP (MV-s-NAP) and showed that it exhibits anti-tumor and immunostimulatory activity in human breast cancer xenograft models. In this study, we utilized a measles-infection-permissive mouse model (transgenic IFNAR KO-CD46Ge) to evaluate the biodistribution and safety of MV-s-NAP. The primary objective was to identify potential toxic side effects and confirm the safety of the proposed clinical doses of MV-s-NAP prior to a phase I clinical trial of intratumoral administration of MV-s-NAP in patients with MBC. Both subcutaneous delivery (corresponding to the clinical trial intratumoral administration route) and intravenous (worst case scenario) delivery of MV-s-NAP were well tolerated: no significant clinical, laboratory or histologic toxicity was observed. This outcome supports the safety of MV-s-NAP for oncolytic virotherapy of MBC. The first-in-human clinical trial of MV-s-NAP in patients with MBC (ClinicalTrials.gov: NCT04521764) was subsequently activated.

3.
J Clin Invest ; 131(13)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34196308

RESUMO

Clinical immunotherapy approaches are lacking efficacy in the treatment of glioblastoma (GBM). In this study, we sought to reverse local and systemic GBM-induced immunosuppression using the Helicobacter pylori neutrophil-activating protein (NAP), a potent TLR2 agonist, as an immunostimulatory transgene expressed in an oncolytic measles virus (MV) platform, retargeted to allow viral entry through the urokinase-type plasminogen activator receptor (uPAR). While single-agent murine anti-PD1 treatment or repeat in situ immunization with MV-s-NAP-uPA provided modest survival benefit in MV-resistant syngeneic GBM models, the combination treatment led to synergy with a cure rate of 80% in mice bearing intracranial GL261 tumors and 72% in mice with CT-2A tumors. Combination NAP-immunovirotherapy induced massive influx of lymphoid cells in mouse brain, with CD8+ T cell predominance; therapeutic efficacy was CD8+ T cell dependent. Inhibition of the IFN response pathway using the JAK1/JAK2 inhibitor ruxolitinib decreased PD-L1 expression on myeloid-derived suppressor cells in the brain and further potentiated the therapeutic effect of MV-s-NAP-uPA and anti-PD1. Our findings support the notion that MV strains armed with bacterial immunostimulatory antigens represent an effective strategy to overcome the limited efficacy of immune checkpoint inhibitor-based therapies in GBM, creating a promising translational strategy for this lethal brain tumor.


Assuntos
Antígenos de Bactérias/uso terapêutico , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Terapia Viral Oncolítica/métodos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/uso terapêutico , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linfócitos T CD8-Positivos/imunologia , Morte Celular/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Citocinas/metabolismo , Efeito Citopatogênico Viral , Feminino , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Pesquisa Translacional Biomédica , Internalização do Vírus
4.
Mol Ther Oncolytics ; 19: 136-148, 2020 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-33145397

RESUMO

Measles virus (MV) Edmonston derivative strains are attractive vector platforms in vaccine development and oncolytic virotherapy. Helicobacter pylori heat shock protein A (HspA) is a bacterial heat shock chaperone with essential function as a Ni-ion scavenging protein. We generated and characterized the immunogenicity of an attenuated MV strain encoding the HspA transgene (MV-HspA). MV-HspA showed faster replication within 48 h of infection with >10-fold higher titers and faster accumulation of the MV proteins. It also demonstrated a superior tumor-killing effect in vitro against a variety of human solid tumor cell lines, including sarcoma, ovarian and breast cancer. Two intraperitoneal (i.p.) doses of 106 50% tissue culture infectious dose (TCID50) MV-HspA significantly improved survival in an ovarian cancer xenograft model: 63.5 days versus 27 days for the control group. The HspA transgene induced a humoral immune response in measles-permissive Ifnarko-CD46Ge transgenic mice. Eight of nine animals developed a long-term anti-HspA antibody response with titers of 1:400 to 1:12,800 without any negative impact on development of protective anti-MV immune memory. MV-HspA triggered an immunogenic cytopathic effect as measured by an HMGB1 assay. The absence of significant elevation of PD-L1 expression indicated that vector-encoded HspA could act as an immunomodulator on the immune check point axis. These data demonstrate that MV-HspA is a potent oncolytic agent and vaccine candidate for clinical translation in cancer treatment and immunoprophylaxis against H. pylori.

5.
Virus Res ; 263: 145-150, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30684519

RESUMO

Measles virus (MV), a paramyxovirus, is one of the most contagious human pathogens and is responsible for thousands of deaths annually. Wild-type MV evolved to counter the innate immune system by avoiding both type I interferon (IFN) induction and inhibiting IFN signaling through the JAK/STAT pathway. However, virus replication is significantly inhibited in IFN-pretreated cells. Similarly, MV vaccine derived strains are inhibited by IFN pretreatment, but vaccine strains also induce IFN. Despite the significant progress in understanding the interactions between MV and the IFN pathway, the IFN stimulated genes (ISGs) that inhibit MV replication remain largely unknown. The aim of this study is to identify specific ISGs that mediate restriction of MV. In this study, we report that Radical S-adenosyl methionine domain containing 2 (RSAD2) restricts MV infection at the stage of virus release in infected 293T cells. Furthermore, attenuated MV strains are currently being developed as a novel treatment for solid and hematological malignancies. Therefore, we tested the impact of RSAD2 expression in an oncolytic virotherapy context using a MV permissive ovarian cancer line (SR-B2). As measured in 293T cells, MV release was also impaired in SR-B2 cells transduced to express RSAD2 in vitro. Additionally, oncolytic MV therapeutic efficacy was impaired in SR-B2 cells transduced to express RSAD2 in vivo. Overall, we identify RSAD2 as a novel restriction factor for MV by inhibiting the release of virus. These results provide important information regarding the interaction between MV and the innate immune system, as well as implications for the design of oncolytic MV platforms.


Assuntos
Interações Hospedeiro-Patógeno , Imunidade Inata , Vírus do Sarampo/imunologia , Vírus do Sarampo/fisiologia , Proteínas/metabolismo , Liberação de Vírus , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/virologia , Humanos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH
6.
J Natl Cancer Inst ; 110(10): 1123-1132, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29788332

RESUMO

Background: Attenuated measles virus (MV) strains are promising agents currently being tested against solid tumors or hematologic malignancies in ongoing phase I and II clinical trials; factors determining oncolytic virotherapy success remain poorly understood, however. Methods: We performed RNA sequencing and gene set enrichment analysis to identify pathways differentially activated in MV-resistant (n = 3) and -permissive (n = 2) tumors derived from resected human glioblastoma (GBM) specimens and propagated as xenografts (PDX). Using a unique gene signature we identified, we generated a diagonal linear discriminant analysis (DLDA) classification algorithm to predict MV responders and nonresponders, which was validated in additional randomly selected GBM and ovarian cancer PDX and 10 GBM patients treated with MV in a phase I trial. GBM PDX lines were also treated with the US Food and Drug Administration-approved JAK inhibitor, ruxolitinib, for 48 hours prior to MV infection and virus production, STAT1/3 signaling and interferon stimulated gene expression was assessed. All statistical tests were two-sided. Results: Constitutive interferon pathway activation, as reflected in the DLDA algorithm, was identified as the key determinant for MV replication, independent of virus receptor expression, in MV-permissive and -resistant GBM PDXs. Using these lines as the training data for the DLDA algorithm, we confirmed the accuracy of our algorithm in predicting MV response in randomly selected GBM PDX ovarian cancer PDXs. Using the DLDA prediction algorithm, we demonstrate that virus replication in patient tumors is inversely correlated with expression of this resistance gene signature (ρ = -0.717, P = .03). In vitro inhibition of the interferon response pathway with the JAK inhibitor ruxolitinib was able to overcome resistance and increase virus production (1000-fold, P = .03) in GBM PDX lines. Conclusions: These findings document a key mechanism of tumor resistance to oncolytic MV therapy and describe for the first time the development of a prediction algorithm to preselect for oncolytic treatment or combinatorial strategies.


Assuntos
Interferons/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Terapia Viral Oncolítica , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Expressão Gênica , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Vírus do Sarampo/genética , Camundongos , Neoplasias/patologia , Vírus Oncolíticos/genética , Reprodutibilidade dos Testes , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Am J Pathol ; 187(1): 33-41, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27855279

RESUMO

Hereditary tyrosinemia type 1 (HT1) is an autosomal recessive disease caused by deficiency in fumarylacetoacetate hydrolase, the last enzyme in the tyrosine catabolic pathway. In this study, we investigated whether fumarylacetoacetate hydrolase deficient (FAH-/-) pigs, a novel large-animal model of HT1, develop fibrosis and cirrhosis characteristic of the human disease. FAH-/- pigs were treated with the protective drug 2-(2-nitro-4-trifluoromethylbenzoyl)-1, 3 cyclohexandione (NTBC) at a dose of 1 mg/kg per day initially after birth. After 30 days, they were assigned to one of three groups based on dosing of NTBC. Group 1 received ≥0.2 mg/kg per day, group 2 cycled on/off NTBC (0.05 mg/kg per day × 1 week/0 mg/kg per day × 3 weeks), and group 3 received no NTBC thereafter. Pigs were monitored for features of liver disease. Animals in group 1 continued to have weight gain and biochemical analyses comparable to wild-type pigs. Animals in group 2 had significant cessation of weight gain, abnormal biochemical test results, and various grades of fibrosis and cirrhosis. No evidence of hepatocellular carcinoma was detected. Group 3 animals declined rapidly, with acute liver failure. In conclusion, the FAH-/- pig is a large-animal model of HT1 with clinical characteristics that resemble the human phenotype. Under conditions of low-dose NTBC, FAH-/- pigs developed liver fibrosis and portal hypertension, and thus may serve as a large-animal model of chronic liver disease.


Assuntos
Tirosinemias/patologia , Animais , Doença Crônica , Modelos Animais de Doenças , Técnicas de Imagem por Elasticidade , Feminino , Heptanoatos/metabolismo , Humanos , Hidrolases/deficiência , Hidrolases/metabolismo , Rim/metabolismo , Rim/patologia , Fígado/patologia , Fígado/fisiopatologia , Cirrose Hepática/patologia , Espectroscopia de Ressonância Magnética , Masculino , Redes e Vias Metabólicas , Fenótipo , Pressão na Veia Porta , Sus scrofa , Tirosina/metabolismo , Aumento de Peso
8.
Vaccine ; 31(42): 4795-801, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23948230

RESUMO

Helicobacter pylori neutrophil-activating protein (NAP) is a toll-like receptor 2 (TLR2) agonist and potent immunomodulator inducing Th1-type immune response. Here we present data about characterization of the humoral immune response against NAP-tagged antigens, encoded by attenuated measles virus (MV) vector platform, in MV infection susceptible type I interferon receptor knockout and human CD46 transgenic (Ifnarko-CD46Ge) mice. Immunogenicity of MV expressing a full-length human immunoglobulin lambda light chain (MV-lambda) was compared to that of MV expressing lambda-NAP chimeric protein (MV-lambda-NAP). MV-lambda-NAP immunized Ifnarko-CD46Ge mice developed significantly higher (6-20-fold) anti-lambda ELISA titers as compared to the MV-lambda-immunized control animal group, indicating that covalently-linked NAP co-expression significantly enhanced lambda immunogenicity. In contrast, ELISA titers against MV antigens were not significantly different between the animals vaccinated with MV-lambda or MV-lambda-NAP. NAP-tagged antigen expression did not affect development of protective anti-measles immunity. Both MV-lambda and MV-lambda-NAP-immunized groups showed strong virus neutralization serum titers in plaque reduction microneutralization test. These results demonstrated that MV-encoded lambda-NAP is highly immunogenic as compared to the unmodified full-length lambda chain. Boost of immune response to poor immunogens using live vectors expressing NAP-tagged chimeric antigens is an attractive approach with potential application in immunoprophylaxis of infectious diseases and cancer immunotherapy.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos/imunologia , Proteínas de Bactérias/imunologia , Portadores de Fármacos , Vírus do Sarampo/imunologia , Adjuvantes Imunológicos/genética , Animais , Anticorpos/sangue , Antígenos/genética , Proteínas de Bactérias/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Vírus do Sarampo/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Testes de Neutralização , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
9.
J Immunol Methods ; 384(1-2): 1-9, 2012 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-22750540

RESUMO

Neutrophil-activating protein (NAP) is a major virulence factor expressed by Helicobacter pylori isolates associated with severe chronic gastroduodenal inflammation and peptic ulcers. NAP is one of the main protective antigens and a target for vaccine development against Helicobacter infection. In addition, NAP is a potent immune stimulator with potential application as a general vaccine adjuvant and in treatment of allergic diseases and cancer immunotherapy. NAP-specific immunoassays are needed for both H. pylori diagnostics and characterization of NAP-based vaccines and immunomodulatory preparations. We generated a panel of NAP-specific monoclonal antibodies (MAbs) by immunization of BALB/c mice with synthetic NAP peptides. The antibody reactivity against recombinant or native NAP antigen was characterized by enzyme-linked immunosorbent assay (ELISA), immunoblotting and immunofluorescence. A sensitive capture ELISA was developed using MAbs 23C8 and 16F4 (directed against different NAP epitopes) for detection of native or measles virus (MV) vector-expressed recombinant NAP in a concentration range of 4 ng/ml to 2000 ng/ml. MAb 23C8 antigen-binding depends on Tyr101 in a variable amino acid sequence of the NAP molecule, indicating the existence of antigenic variants among H. pylori strains. MAb 16F4 reacted with NAP from different H. pylori strains and was a sensitive tool for detection of small amounts of isolated NAP antigen or whole bacteria by immunoblotting or immunofluorescence. In conclusion, MAb-based immunoassays are highly specific and sensitive for detection of native NAP antigen and recombinant NAP immunostimulatory transgenes expressed by replication competent virus vectors.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Chlorocebus aethiops , Mapeamento de Epitopos , Epitopos/genética , Epitopos/imunologia , Epitopos/metabolismo , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Helicobacter pylori/genética , Helicobacter pylori/imunologia , Helicobacter pylori/metabolismo , Immunoblotting , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Células Vero
10.
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
11.
Methods Mol Biol ; 797: 141-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21948475

RESUMO

Despite significant advances in recent years, treatment of metastatic malignancies remains a significant challenge. There is an urgent need for development of novel therapeutic approaches. Virotherapy approaches have considerable potential, and among them measles virus (MV) vaccine strains have emerged as a promising oncolytic platform. Retargeted MV strains deriving from the Edmonston vaccine lineage (MV-Edm) have shown comparable antitumor efficacy to unmodified strains against receptor expressing tumor cells with improved therapeutic index. Here, we describe the construction, rescue, amplification, and titration of fully retargeted MV-Edm derivatives displaying tumor specific receptor binding ligands on the viral surface in combination with H protein CD46 and SLAM entry ablating mutations.


Assuntos
Vírus do Sarampo/genética , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Vetores Genéticos , Humanos , Ligantes , Vacina contra Sarampo , Camundongos , Camundongos Nus , Metástase Neoplásica , Células Vero , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Vaccine ; 29(8): 1710-20, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21182995

RESUMO

Helicobacter pylori is a Gram-negative, spiral-shaped microorganism associated with acute and chronic gastritis, peptic ulcer, gastric cancer and gastric lymphomas in humans. H. pylori neutrophil-activating protein (NAP) is a major virulence factor playing a central role in pathogenesis of mucosal inflammation by immune cell attraction and Th1 cytokine response polarization. NAP is protective antigen and promising vaccine candidate against H. pylori infection. Here we present the development of measles virus (MV) vaccine strain encoding the NAP antigen. In order to facilitate the extracellular transport and detection, NAP was inserted in the human lambda immunoglobulin chain replacing a major part of the variable domain. We generated two MV vectors expressing secretory NAP forms: MV-lambda-NAP encoding the full-length constant lambda light chain domain and MV-s-NAP encoding only the N-terminus of the lambda light chain with the leader peptide. Immunization of MV permissive Ifnarko-CD46Ge transgenic mice by a single intraperitoneal injection of the NAP-expressing strains induced a robust, long-term humoral and cellular immune response against MV. Nine months post vaccination measles-neutralizing antibody titers were above the serum level considered protective for humans. Furthermore, all animals immunized with MV strains expressing the secretory NAP antigen developed strong humoral immunity against NAP, reaching titers >1:10,000 within 2-4 weeks. IFN-γ ELISpot assay confirmed that NAP-encoding MV vectors can also stimulate NAP-specific cell-mediated immunity. Our data demonstrate that MV is an excellent vector platform for expression of bacterial antigens and development of vaccines for H. pylori immunoprophylaxis in humans.


Assuntos
Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Infecções por Helicobacter/prevenção & controle , Vírus do Sarampo/imunologia , Sarampo/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Humanos , Imunidade Humoral , Interferon gama/imunologia , Interleucina-8/imunologia , Sarampo/imunologia , Vírus do Sarampo/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Testes de Neutralização , Proteínas Recombinantes/imunologia , Vacinas Atenuadas/imunologia , Células Vero
13.
Breast Cancer Res Treat ; 122(3): 745-54, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19894113

RESUMO

Breast cancer is the second leading cause of malignant effusions in cancer patients. Pleural effusion indicates incurable disease with limited palliative treatment options and poor outcome. Here, we demonstrate the therapeutic efficacy of measles virus (MV) vaccine strain derivative against malignant pleural effusion in an MDA-MB-231 xenograft model of advanced breast cancer. Both systemic intravenous (i.v.) and intrapleural (t.t.) administered virus caused massive infection and syncytia formation in the pleural tumor deposits. Intrapleural administration of 1.5 x 10(6) plaque-forming units (PFU) total dose of MV significantly improved median survival by approximately 80% compared to the control animal group. Furthermore, we tested human dendritic cells as carriers for delivery of oncolytic MV infection to breast cancer pleural metastases. Carrier-delivered MV infection prevented accumulation of the pleural exudate and also significantly improved the survival of the treated mice. This is the first demonstration of the therapeutic potential of oncolytic virotherapy against malignant pleural effusions in a pre-clinical model of advanced breast cancer.


Assuntos
Neoplasias da Mama/terapia , Modelos Animais de Doenças , Vacina contra Sarampo/uso terapêutico , Vírus do Sarampo/imunologia , Terapia Viral Oncolítica , Derrame Pleural Maligno/terapia , Animais , Apoptose/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Chlorocebus aethiops , Células Dendríticas/imunologia , Feminino , Humanos , Sarampo/complicações , Sarampo/imunologia , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Vírus do Sarampo/genética , Camundongos , Camundongos Nus , Derrame Pleural Maligno/genética , Derrame Pleural Maligno/imunologia , Taxa de Sobrevida , Células Tumorais Cultivadas , Células Vero , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Mol Ther ; 17(12): 2041-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19773744

RESUMO

Prostate cancer cells overexpress the measles virus (MV) receptor CD46. Herein, we evaluated the antitumor activity of an oncolytic derivative of the MV Edmonston (MV-Edm) vaccine strain engineered to express the human sodium iodide symporter (NIS; MV-NIS virus). MV-NIS showed significant cytopathic effect (CPE) against prostate cancer cell lines in vitro. Infected cells effectively concentrated radioiodide isotopes as measured in vitro by Iodide-125 ((125)I) uptake assays. Virus localization and spread in vivo could be effectively followed by imaging of (123)I uptake. In vivo administration of MV-NIS either locally or systemically (total dose of 9 x 10(6) TCID(50)) resulted in significant tumor regression (P < 0.05) and prolongation of survival (P < 0.01). Administration of (131)I further enhanced the antitumor effect of MV-NIS virotherapy (P < 0.05). In conclusion, MV-NIS is an oncolytic vector with significant antitumor activity against prostate cancer, which can be further enhanced by (131)I administration. The NIS transgene allows viral localization and monitoring by noninvasive imaging which can facilitate dose optimization in a clinical setting.


Assuntos
Diagnóstico por Imagem , Radioisótopos do Iodo/metabolismo , Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/terapia , Simportadores/genética , Animais , Proliferação de Células , Chlorocebus aethiops , Efeito Citopatogênico Viral , Engenharia Genética , Humanos , Radioisótopos do Iodo/uso terapêutico , Masculino , Vírus do Sarampo/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/patologia , Simportadores/metabolismo , Células Tumorais Cultivadas , Raios Ultravioleta , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Mol Ther ; 17(8): 1395-403, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19471250

RESUMO

Preferential killing of transformed cells, while keeping normal cells and organs unharmed, is the main goal of cancer gene therapy. Genetically engineered trackable markers and imaging reporters enable noninvasive monitoring of transduction efficiency and pharmacokinetics of anticancer virotherapeutics. However, none of these reporters can differentiate between infection in the targeted tumors and that in the normal tissue. Thus, we constructed oncolytic measles virus (MV) armed with a human light immunoglobulin chain reporter gene for the treatment of multiple myeloma (MM). Excessive production of monoclonal immunoglobulin is a key characteristic and marker for diagnostics of MM. Once expressed in infected target cells, vector-encoded lambda protein recombines with myeloma IgG-kappa immunoglobulin creating a unique IgG-kappa/lambda. A modified immunoassay technique allows precise quantification of converted marker molecules. Only antibody producing cells were able to assemble this chimeric immunoglobulin molecule, whereas other cells secreted only free lambda light chain. Human myeloma xenografts inoculated with lambda chain expressing MV secreted converted IgG-kappa/lambda in the plasma of tumor bearing animals and elevated reporter levels correlated with response to the therapy. This is the first report of a gene therapy vector engineered to discriminate between infection in malignant and normal cells by molecular modification of a tumor-specific protein.


Assuntos
Biomarcadores Tumorais/metabolismo , Cadeias Leves de Imunoglobulina/metabolismo , Vírus do Sarampo/fisiologia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica/métodos , Animais , Biomarcadores Tumorais/genética , Linhagem Celular , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Cadeias Leves de Imunoglobulina/genética , Imuno-Histoquímica , Vírus do Sarampo/genética , Camundongos , Camundongos SCID , Vírus Oncolíticos , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Prostate ; 69(1): 82-91, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18973133

RESUMO

BACKGROUND: No curative therapy is currently available for locally advanced or metastatic prostate cancer. Oncolytic viruses represent a novel class of therapeutic agents that demonstrates no cross-resistance with existing approaches and can therefore be combined with conventional treatment modalities. Measles virus strains deriving from the Edmonston (MV-Edm) vaccine strain have shown considerable oncolytic activity against a variety of solid tumers and hematologic malignancies. In this study, we investigated the antitumor potential of recombinant MV-Edm derivatives as novel oncolytic agents against prostate cancer. METHODS: The susceptibility of prostate cancer cell lines (PC-3, DU-145, and LNCaP) to measles virus infection was demonstrated using an MV-Edm derivative expressing green fluorescent protein (GFP). MV-Edm replication in prostate cancer cell lines was assessed by one step viral growth curves. The oncolytic effect of an MV-Edm strain engineered to express the human carcinoembryonic antigen (CEA) was demonstrated in vitro by MTT assays and in vivo in subcutaneous PC-3 xenografts. CEA levels were quantitated in cell supernatants and mouse serum samples. RESULTS: Recombinant MV-Edm strains can effectively infect, replicate in and kill prostate cancer cells. Intratumoral administration of MV-CEA at a total dose of 6 x 10(6) TCID50 resulted in statistically significant tumor growth delay (P = 0.004) and prolongation of survival (P = 0.001) in a subcutaneous PC-3 xenograft model. Viral growth kinetics paralleled CEA production. CONCLUSIONS: MV-CEA has potent antitumor activity against prostate cancer cell lines and xenografts. Viral gene expression during treatment can be determined by monitoring of CEA levels in the serum; the latter could allow dose optimization and tailoring of individualized treatment protocols.


Assuntos
Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/terapia , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Citometria de Fluxo , Engenharia Genética/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Injeções Intravenosas , Luciferases/genética , Masculino , Vírus do Sarampo/imunologia , Proteína Cofatora de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/patologia , Proteínas Recombinantes de Fusão/genética , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
17.
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
18.
Mol Ther ; 15(1): 114-22, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17164782

RESUMO

Attenuated measles viruses (MVs) propagate selectively in human tumor cells, and phase I clinical trials are currently underway to test their oncolytic activity. A major theoretical impediment to systemic MV application is the presence of pre-existing antiviral immunity. We hypothesized that autologous MV-infected cells might be a more reliable vehicle than cell-free virions to deliver the infection to tumor cells in subjects with neutralizing titers of anti-measles antibodies. Our in vitro studies, using a dual-color fluorescent model, demonstrated efficient cell-to-cell transfer of infection via heterofusion. In contrast to infection by naked virions, heterofusion between infected cell carriers and tumor cells was more resistant to antibody neutralization. Infected monocytic, endothelial, or stimulated peripheral blood cells could deliver oncolytic MV to tumor lesions in vivo, after intravenous (i.v.) or intraperitoneal (i.p.) administration. Single or repeated i.p. injections of monocytic carriers significantly improved survival of animals bearing human ovarian cancer xenografts. Systemic or i.p. injection of MV-infected cells successfully transferred infection by heterofusion to Raji lymphomas or hepatocellular carcinoma tumors in the presence of neutralizing antibodies. These results suggest a novel strategy for systemic delivery of oncolytic virotherapy in cancer patients that can "bypass" the pre-existing humoral immunity against MV.


Assuntos
Sarampo , Morbillivirus/fisiologia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Transgenes/genética , Internalização do Vírus , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Expressão Gênica , Terapia Genética , Humanos , Injeções Intravenosas , Camundongos , Camundongos SCID , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/terapia , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Virol ; 80(17): 8530-40, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16912303

RESUMO

Antibodies to viral surface glycoproteins play a crucial role in immunity to measles by blocking both virus attachment and subsequent fusion with the host cell membrane. Here, we demonstrate that certain immunoglobulin G (IgG) antibodies can also enhance the entry of measles virus (MV) into monocytes and macrophages. Antibody-dependent enhancement of infectivity was observed in mouse and human macrophages using virions opsonized by a murine monoclonal antibody against the MV hemagglutinin (H) glycoprotein, polyclonal mouse anti-MV IgG, or diluted measles-immune human sera. Neither H-specific Fab fragments nor H-specific IgM could enhance MV entry in monocytes or macrophages, indicating involvement of a Fc gamma receptor (FcgammaR)-mediated mechanism. Preincubation with an anti-fusion protein (anti-F) monoclonal antibody or a fusion-inhibitory peptide blocked infection, indicating that a functional F protein was required for viral internalization. Classical complement pathway activation did not promote infection through complement receptors and inhibited anti-H IgG-mediated enhancement. In vivo, antibody-enhanced infection allowed MV to overcome a highly protective systemic immune response in preimmunized IfnarKo-Ge46 transgenic mice. These data demonstrate a previously unidentified mechanism that may contribute to morbillivirus pathogenesis where H-specific IgG antibodies promote the spread of MV infection among FcgammaR-expressing host cells. The findings point to a new model for the pathogenesis of atypical MV infection observed after immunization with formalin-inactivated MV vaccine and underscore the importance of the anti-F response after vaccination.


Assuntos
Anticorpos Antivirais/imunologia , Anticorpos Facilitadores , Imunoglobulina G/imunologia , Vírus do Sarampo/patogenicidade , Sarampo/imunologia , Animais , Linhagem Celular , Hemaglutininas Virais/imunologia , Humanos , Imunização , Macrófagos/virologia , Sarampo/fisiopatologia , Sarampo/prevenção & controle , Sarampo/virologia , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/imunologia , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia , Camundongos , Camundongos Transgênicos , Monócitos/virologia , Células U937/virologia , Proteínas Virais de Fusão/imunologia
20.
Microbes Infect ; 6(10): 901-10, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15310466

RESUMO

Protective properties of immunoglobulin A (IgA) monoclonal antibodies (MAbs) directed against O and H antigens of Salmonella enterica serotype Enteritidis (S. enteritidis) were evaluated in a model of generalized infection after intranasal (i.n.) inoculation of BALB/c mice. Passive i.n. instillation of antibodies 1 h before i.n. challenge did not prevent infection, and mice developed rapid inflammatory response in the lower respiratory tract. The passive systemic immunization was partially protective and a single intravenous (i.v.) injection of both O and H antigen specific IgA antibodies prolonged survival period of the infected animals. Permanent secretion of O:9 specific IgA MAb 177E6 into the respiratory tract in a "backpack" tumor model protected 50% of animals infected i.n. with a high dose of virulent S. enteritidis strain. Thus, secretory IgA (S-IgA) directed against O:9 antigen alone can prevent bacterial invasion in the respiratory epithelium.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Bactérias/imunologia , Imunoglobulina A/imunologia , Pneumopatias/microbiologia , Antígenos O/imunologia , Salmonelose Animal/imunologia , Salmonella enteritidis/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/farmacologia , Anticorpos Monoclonais/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Imunização Passiva/métodos , Imunoglobulina A/farmacologia , Cinética , Pneumopatias/imunologia , Pneumopatias/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Salmonelose Animal/microbiologia , Salmonelose Animal/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA