Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 187(1): 33-41, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27855279

RESUMEN

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.


Asunto(s)
Tirosinemias/patología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Diagnóstico por Imagen de Elasticidad , Femenino , Heptanoatos/metabolismo , Humanos , Hidrolasas/deficiencia , Hidrolasas/metabolismo , Riñón/metabolismo , Riñón/patología , Hígado/patología , Hígado/fisiopatología , Cirrosis Hepática/patología , Espectroscopía de Resonancia Magnética , Masculino , Redes y Vías Metabólicas , Fenotipo , Presión Portal , Sus scrofa , Tirosina/metabolismo , Aumento de Peso
2.
J Virol ; 88(24): 14161-71, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25275122

RESUMEN

UNLABELLED: Many viruses utilize cell adhesion molecules of the immunoglobulin superfamily as receptors. In particular, viruses of different classes exploit nectins. The large DNA viruses, herpes simplex and pseudorabies viruses, use ubiquitous nectins 1 and 2. The negative-strand RNA virus measles virus (MeV) uses tissue-specific nectin-4, and the positive-strand RNA virus poliovirus uses nectin-like 5 (necl-5), also known as poliovirus receptor. These viruses contact the BC, C'C", and FG loops on the upper tip of their receptor's most membrane-distal domain. This location corresponds to the newly defined canonical adhesive interface of nectins, but how viruses utilize this interface has remained unclear. Here we show that the same key residues in the BC and FG loops of nectin-4 govern binding to the MeV attachment protein hemagglutinin (H) and cell entry, nectin-4 homodimerization, and heterodimerization with nectin-1. On the other hand, residues in the C'C" loop necessary for homo- and heterotypic interactions are dispensable for MeV-induced fusion and cell entry. Remarkably, the C'C" loop governs dissociation of the nectin-4 and H ectodomains. We provide formal proof that H can interfere with the formation of stable nectin-1/nectin-4 heterodimers. Finally, while developing an alternative model to study MeV spread, we observed that polarized primary pig airway epithelial sheets cannot be infected. We show that a single amino acid variant in the BC loop of pig nectin-4 fully accounts for restricted MeV entry. Thus, the three loops forming the adhesive interface of nectin-4 have different roles in supporting MeV H association and dissociation and MeV-induced fusion. IMPORTANCE: Different viruses utilize nectins as receptors. Nectins are immunoglobulin superfamily glycoproteins that mediate cell-cell adhesion in vertebrate tissues. They interact through an adhesive interface located at the top of their membrane-distal domain. How viruses utilize the three loops forming this interface has remained unclear. We demonstrate that while nectin-nectin interactions require residues in all three loops, the association of nectin-4 with the measles virus hemagglutinin requires only the BC and FG loops. However, we discovered that residues in the C'C" loop modulate the dissociation of nectin-4 from the viral hemagglutinin. Analogous mechanisms may support cell entry of other viruses that utilize nectins or other cell adhesion molecules of the immunoglobulin superfamily as receptors.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hemaglutininas Virales/metabolismo , Virus del Sarampión/fisiología , Multimerización de Proteína , Receptores Virales/metabolismo , Acoplamiento Viral , Secuencia de Aminoácidos , Animales , Línea Celular , Humanos , Datos de Secuencia Molecular , Nectinas , Alineación de Secuencia , Internalización del Virus
3.
Mol Diagn Ther ; 28(2): 153-168, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38150172

RESUMEN

Development of successful cancer therapeutics requires exploration of the differences in genetics, metabolism, and interactions with the immune system among malignant and normal cells. The clinical observation of spontaneous tumor regression following natural infection with microorganism has created the premise of their use as cancer therapeutics. Oncolytic viruses (OVs) originate from viruses with attenuated virulence in humans, well-characterized vaccine strains of known human pathogens, or engineered replication-deficient viral vectors. Their selectivity is based on receptor expression level and post entry restriction factors that favor replication in the tumor, while keeping the normal cells unharmed. Clinical trials have demonstrated a wide range of patient responses to virotherapy, with subgroups of patients significantly benefiting from OV administration. Tumor-specific gene signatures, including antiviral interferon-stimulated gene (ISG) expression profile, have demonstrated a strong correlation with tumor permissiveness to infection. Furthermore, the combination of OVs with immunotherapeutics, including anticancer vaccines and immune checkpoint inhibitors [ICIs, such as anti-PD-1/PD-L1 or anti-CTLA-4 and chimeric antigen receptor (CAR)-T or CAR-NK cells], could synergistically improve the therapeutic outcome. Creating response prediction algorithms represents an important step for the transition to individualized immunovirotherapy approaches in the clinic. Integrative predictors could include tumor mutational burden (TMB), inflammatory gene signature, phenotype of tumor-infiltrating lymphocytes, tumor microenvironment (TME), and immune checkpoint receptor expression on both immune and target cells. Additionally, the gut microbiota has recently been recognized as a systemic immunomodulatory factor and could further be used in the optimization of individualized immunovirotherapy algorithms.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Virus Oncolíticos/genética , Neoplasias/genética , Neoplasias/terapia , Linfocitos Infiltrantes de Tumor , Inmunomodulación , Microambiente Tumoral/genética
4.
Nat Commun ; 15(1): 493, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38216554

RESUMEN

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.


Asunto(s)
Glioblastoma , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Virus del Sarampión/genética , Antígeno Carcinoembrionario/genética , Recurrencia Local de Neoplasia/terapia , Vacuna Antisarampión , Microambiente Tumoral
5.
Mol Ther ; 20(6): 1139-47, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22334023

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/genética , Neoplasias de la Mama/terapia , Helicobacter pylori , Factores Inmunológicos/genética , Virus del Sarampión/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Orden Génico , Helicobacter pylori/inmunología , Humanos , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Ratones Desnudos , Ratones Transgénicos , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Derrame Pleural Maligno/inmunología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Ther Methods Clin Dev ; 26: 532-546, 2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36092362

RESUMEN

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.

7.
J Immunol Methods ; 492: 112996, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33582147

RESUMEN

Dried blood spots (DBS) are routinely used in screening newborns for treatable disorders. Immunoglobulin extraction from DBS, serum or other biological fluids loaded on filter paper cards could represent a valuable method of specimen preservation in monitoring immune response against pathogens as well as vaccination efficiency. In this study using different sources including serum, and monoclonal antibodies we established parameters for antibody extraction from the filter cards to assess antibody reactivity against Helicobacter pylori, measles virus (MV) and the novel coronavirus SARS-CoV-2 antigens. We demonstrated that DBS and dried undiluted serum result in completely preserved antibody activity for immunoassays, including in virus neutralization assays against MV. Extraction efficiency was determined by IgG concentration measurements. The plaque-reduction neutralization titer 50% of dried human serum spots remained stable after more than 10-day storage - 1:359 vs. 1:345 for the corresponding frozen sample. DBSs could be used to monitor immune response to bacterial and viral antigens following natural exposure or immunization. Mice immunized with recombinant spike protein receptor-binding domain of SARS-CoV-2 developed a strong antibody response by day 14 and reached titers above 1:64,000 on day 21 following the secondary boost immunization as measured on DBS samples in antigen-mediated ELISA. Variability in IgG concentration of eluted DBS could be influenced by factors involved in sample application, extraction process and sample characteristics. Adjustment of antibody specific activity to the eluted IgG concentration can increase accuracy of the result interpretation, including in SARS-CoV-2 serological diagnostics.


Asunto(s)
Anticuerpos Antivirales/aislamiento & purificación , COVID-19/diagnóstico , Pruebas con Sangre Seca , Infecciones por Helicobacter/diagnóstico , Helicobacter pylori/fisiología , Inmunoensayo/métodos , Virus del Sarampión/fisiología , Sarampión/diagnóstico , Monitorización Inmunológica/métodos , SARS-CoV-2/fisiología , Animales , Anticuerpos Monoclonales , Formación de Anticuerpos , Pruebas con Sangre Seca/métodos , Femenino , Humanos , Inmunidad Humoral , Ratones , Ratones Endogámicos BALB C , Pruebas Serológicas , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunación
8.
J Clin Invest ; 131(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34196308

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/uso terapéutico , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Viroterapia Oncolítica/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/uso terapéutico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Linfocitos T CD8-positivos/inmunología , Muerte Celular/inmunología , Línea Celular Tumoral , Terapia Combinada , Citocinas/metabolismo , Efecto Citopatogénico Viral , Femenino , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Virus del Sarampión/genética , Virus del Sarampión/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Investigación Biomédica Traslacional , Internalización del Virus
9.
Breast Cancer Res Treat ; 122(3): 745-54, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19894113

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/terapia , Modelos Animales de Enfermedad , Vacuna Antisarampión/uso terapéutico , Virus del Sarampión/inmunología , Viroterapia Oncolítica , Derrame Pleural Maligno/terapia , Animales , Apoptosis/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Chlorocebus aethiops , Células Dendríticas/inmunología , Femenino , Humanos , Sarampión/complicaciones , Sarampión/inmunología , Vacuna Antisarampión/genética , Vacuna Antisarampión/inmunología , Virus del Sarampión/genética , Ratones , Ratones Desnudos , Derrame Pleural Maligno/genética , Derrame Pleural Maligno/inmunología , Tasa de Supervivencia , Células Tumorales Cultivadas , Células Vero , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Ther ; 17(8): 1395-403, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19471250

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Cadenas Ligeras de Inmunoglobulina/metabolismo , Virus del Sarampión/fisiología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/terapia , Viroterapia Oncolítica/métodos , Animales , Biomarcadores de Tumor/genética , Línea Celular , Chlorocebus aethiops , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Cadenas Ligeras de Inmunoglobulina/genética , Inmunohistoquímica , Virus del Sarampión/genética , Ratones , Ratones SCID , Virus Oncolíticos , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Ther ; 17(12): 2041-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19773744

RESUMEN

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.


Asunto(s)
Diagnóstico por Imagen , Radioisótopos de Yodo/metabolismo , Vacuna Antisarampión/genética , Virus del Sarampión/genética , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Simportadores/genética , Animales , Proliferación Celular , Chlorocebus aethiops , Efecto Citopatogénico Viral , Ingeniería Genética , Humanos , Radioisótopos de Yodo/uso terapéutico , Masculino , Virus del Sarampión/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Próstata/patología , Simportadores/metabolismo , Células Tumorales Cultivadas , Rayos Ultravioleta , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Ther Oncolytics ; 19: 136-148, 2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-33145397

RESUMEN

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.

13.
Prostate ; 69(1): 82-91, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18973133

RESUMEN

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.


Asunto(s)
Vacuna Antisarampión/genética , Virus del Sarampión/genética , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Citometría de Flujo , Ingeniería Genética/métodos , Proteínas Fluorescentes Verdes/genética , Humanos , Inyecciones Intravenosas , Luciferasas/genética , Masculino , Virus del Sarampión/inmunología , Proteína Cofactora de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Próstata/patología , Proteínas Recombinantes de Fusión/genética , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Ther ; 16(9): 1556-1564, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28189011

RESUMEN

The majority of glioblastoma multiforme (GBM) tumors (80%) overexpress interleukin-13 receptor α2 (IL-13Rα2), but there is no expression of IL-13Rα2 in normal brain. Vaccine strains of measles virus have significant antitumor activity against gliomas. We tested the hypothesis that measles virus entry could be retargeted via the IL-13Rα2. MV-GFP-HAA-IL-13 was generated from the Edmonston-NSe vaccine strain, by displaying human IL-13 at the C-terminus of the H protein, and introducing CD46 and signaling lymphocyte activation molecule (SLAM)-ablating mutations in H. The IL-13 retargeted virus showed significant cytopathic effect (CPE) against IL-13Rα2 overexpressing glioma lines, and lack of CPE/viral replication in normal human astrocytes and normal human fibroblasts not expressing IL-13Rα2. In vivo treatment of orthotopically implanted GBM12 xenografts demonstrated significant prolongation of survival in mice treated with the retargeted strain (P < 0.0001), and comparable activity between the IL-13R retargeted strain and MV-GFP (P = 0.6377). In contrast to MV-GFP-treated mice, administration of the retargeted strain in the central nervous system of measles replication-permissive Ifnarko CD46 Ge mice resulted in lack of neurotoxicity. Strains of measles virus retargeted against the glioma-specific IL-13Rα2 receptor have comparable therapeutic efficacy, and improved specificity as compared with the unmodified measles virus strain MV-GFP in vitro and in vivo.

15.
Mol Ther ; 16(9): 1556-64, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18665158

RESUMEN

The majority of glioblastoma multiforme (GBM) tumors (80%) overexpress interleukin-13 receptor alpha2 (IL-13Ralpha2), but there is no expression of IL-13Ralpha2 in normal brain. Vaccine strains of measles virus have significant antitumor activity against gliomas. We tested the hypothesis that measles virus entry could be retargeted via the IL-13Ralpha2. MV-GFP-H(AA)-IL-13 was generated from the Edmonston-NSe vaccine strain, by displaying human IL-13 at the C-terminus of the H protein, and introducing CD46 and signaling lymphocyte activation molecule (SLAM)-ablating mutations in H. The IL-13 retargeted virus showed significant cytopathic effect (CPE) against IL-13Ralpha2 overexpressing glioma lines, and lack of CPE/viral replication in normal human astrocytes and normal human fibroblasts not expressing IL-13Ralpha2. In vivo treatment of orthotopically implanted GBM12 xenografts demonstrated significant prolongation of survival in mice treated with the retargeted strain (P < 0.0001), and comparable activity between the IL-13R retargeted strain and MV-GFP (P = 0.6377). In contrast to MV-GFP-treated mice, administration of the retargeted strain in the central nervous system of measles replication-permissive Ifnar(ko) CD46 Ge mice resulted in lack of neurotoxicity. Strains of measles virus retargeted against the glioma-specific IL-13Ralpha2 receptor have comparable therapeutic efficacy, and improved specificity as compared with the unmodified measles virus strain MV-GFP in vitro and in vivo.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética , Glioma/terapia , Subunidad alfa2 del Receptor de Interleucina-13/uso terapéutico , Interleucina-13/metabolismo , Virus del Sarampión/fisiología , Viroterapia Oncolítica , Animales , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/virología , Chlorocebus aethiops , Citometría de Flujo , Glioma/genética , Glioma/inmunología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Humanos , Subunidad alfa2 del Receptor de Interleucina-13/genética , Sarampión/genética , Sarampión/inmunología , Sarampión/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de la Nucleocápside/metabolismo , Células Tumorales Cultivadas , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Virus Res ; 263: 145-150, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30684519

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Virus del Sarampión/inmunología , Virus del Sarampión/fisiología , Proteínas/metabolismo , Liberación del Virus , Línea Celular , Células Epiteliales/inmunología , Células Epiteliales/virología , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH
17.
Mol Ther ; 15(3): 588-97, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17245355

RESUMEN

The innate antiviral responses of tumor cells are often impaired but may still be sufficient to impede the intratumoral spread of an oncolytic virus. Here, we establish that the oncolytic measles virus (MV-eGFP) induces interferon (IFN) production in human myeloma and ovarian cancer cells. In addition, MV gene expression and virus progeny production were inhibited by IFN treatment of these tumor cells. The P gene of wild-type measles virus encodes P/V/C proteins known to antagonize IFN induction and/or response. We therefore engineered MV-eGFP for IFN evasion and more efficient intratumoral spread by arming it with the P gene from wild-type IC-B strain MV, thus generating MV-eGFP-Pwt. The chimeric virus exhibited reduced IFN sensitivity and diminished capacity to induce IFN in BJAB lymphoma, ARH-77 myeloma cells, and activated peripheral blood mononuclear cells. Interestingly, unlike the wild-type MV, MV-eGFP-Pwt was unable to shut down IFN induction completely. In immunocompromised mice bearing human myeloma xenografts, intravenously administered MV-eGFP-Pwt showed significantly enhanced oncolytic potency compared to MV-eGFP. These results indicate that oncolytic viruses are subject to control by the innate immune defenses of human tumor cells and may therefore be more effective if their natural ability to combat innate immunity is maintained.


Asunto(s)
Inmunidad Innata/inmunología , Virus del Sarampión/genética , Virus Oncolíticos/genética , Animales , Línea Celular Tumoral , Supervivencia Celular , Genes Reporteros/genética , Ingeniería Genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Inyecciones Intravenosas , Interferones/biosíntesis , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Sensibilidad y Especificidad , Proteínas Virales/genética , Proteínas Virales/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Ther ; 15(1): 114-22, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17164782

RESUMEN

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.


Asunto(s)
Sarampión , Morbillivirus/fisiología , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Transgenes/genética , Internalización del Virus , Animales , Anticuerpos Antivirales/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Terapia Genética , Humanos , Inyecciones Intravenosas , Ratones , Ratones SCID , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Natl Cancer Inst ; 110(10): 1123-1132, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29788332

RESUMEN

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.


Asunto(s)
Interferones/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Viroterapia Oncolítica , Transducción de Señal , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Virus del Sarampión/genética , Ratones , Neoplasias/patología , Virus Oncolíticos/genética , Reproducibilidad de los Resultados , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Oncotarget ; 8(53): 91803-91816, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29207686

RESUMEN

Although the majority of breast cancers initially respond to the cytotoxic effects of chemotherapeutic agents, most breast cancer patients experience tumor relapse and ultimately die because of drug resistance. Breast cancer cells undergoing epithelial to mesenchymal transition (EMT) acquire a CD44+/CD24-/ALDH1+ cancer stem cell-like phenotype characterized by an increased capacity for tumor self-renewal, intrinsic drug resistance and high proclivity to develop distant metastases. We uncovered in human breast tumor xenografts a novel non-mitotic role of Aurora-A kinase in promoting breast cancer metastases through activation of EMT and expansion of breast tumor initiating cells (BTICs). In this study we characterized the role of the Aurora-A/SMAD5 oncogenic axis in the induction of chemoresistance. Breast cancer cells overexpressing Aurora-A showed resistance to conventional chemotherapeutic agents, while treatment with alisertib, a selective Aurora-A kinase inhibitor, restored chemosensitivity. Significantly, SMAD5 expression was required to induce chemoresistance and maintain a breast cancer stem cell-like phenotype, indicating that the Aurora-A/SMAD5 oncogenic axis promotes chemoresistance through activation of stemness signaling. Taken together, these findings identified a novel mechanism of drug resistance through aberrant activation of the non-canonical Aurora-A/SMAD5 oncogenic axis in breast cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA