Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Virol Methods ; 302: 114487, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35104497

RESUMEN

The use of oncolytic viruses (OV) to precisely target and eliminate tumors ('virotherapy') is a rapidly evolving therapeutic approach to treating cancer. A major obstacle in virotherapy, especially for systemic administration, is the host's immune response towards the OV. In the case of measles virus (MeV), most individuals have been immunized against this agent leading to pre-existing neutralizing antibodies that can impair OV delivery to the tumor. These antibodies predominantly target the hemagglutinin (H) and fusion (F) envelope glycoproteins displayed at the particle's surface. Here, we introduce a novel and versatile pseudotyping platform for rapid envelope exchange of oncolytic MeV that allows for engineering of chimeric viruses invulnerable to pre-existing anti-MeV antibodies. Using this system, we have successfully exchanged the MeV F and H proteins with the glycoprotein G of vesicular stomatitis virus (VSV) and the surface proteins of Newcastle disease virus (NDV) or canine distemper virus (CDV), all of which are not endemic in the general human population. While the MeV-VSV and MeV-NDV pseudotypes were non-functional, the MeV-CDV pseudotype was successfully propagated to high-titer virus stocks. This study describes the successful generation of a robust envelope exchange platform for oncolytic MeV while also highlighting its intricate pseudotyping tolerance.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Animales , Anticuerpos Neutralizantes , Virus del Sarampión/genética , Virus Oncolíticos/genética , Virus de la Estomatitis Vesicular Indiana
2.
Cancer Gene Ther ; 21(9): 373-80, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25145311

RESUMEN

Precise oncotropism is required for successful systemic administration of next-generation oncolytic measles viruses (MVs). We have previously established a system for efficient post-entry targeting by insertion of synthetic microRNA target sites (miRTS) into the MV genome, thereby repressing replication in the presence of cognate microRNAs. Thus, differential expression of microRNAs, as frequently observed in normal compared with malignant tissues, can be exploited to increase vector specificity and safety. Here we report the combination of miRTS for different microRNAs in a single vector to detarget pivotal organs at risk during systemic administration (liver, brain, gastrointestinal tract). Accordingly, miRTS for miR-122, miR-7 and miR-148a that are enriched in these tissues were inserted to create multi-tissue-detargeted MV (MV-EGFP(mtd)). Replication of MV-EGFP(mtd) is repressed in cell lines as well as in non-transformed primary human hepatocytes and liver slices expressing cognate microRNAs. Oncolytic potency of MV-EGFP(mtd) is retained in a model of pancreatic cancer in vitro and in vivo. This work is a proof-of-concept that favorable expression profiles of multiple microRNAs can be exploited concomitantly to reshape the tropism of MV without compromising oncolytic efficacy. This strategy can be adapted to different vectors and cancer entities for safe and efficient high-dose systemic administration in clinical trials.


Asunto(s)
Vectores Genéticos/genética , Virus del Sarampión/genética , MicroARNs/genética , Virus Oncolíticos/genética , Animales , Secuencia de Bases , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Chlorocebus aethiops , Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Orden Génico , Genes Reporteros , Vectores Genéticos/administración & dosificación , Humanos , Ratones , MicroARNs/química , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Interferencia de ARN , Transducción Genética , Células Vero , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Gynecol Oncol ; 130(2): 362-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23676551

RESUMEN

OBJECTIVE: To preclinical assess the feasibility of combining oncolytic measles vaccine virus (MeV) with suicide gene therapy for ovarian cancer treatment. METHODS: We genetically engineered a recombinant MeV armed with a yeast-derived bifunctional suicide gene that encodes for cytosine deaminase and uracil phosphoribosyltransferase (MeV-SCD). From this suicide gene, a chimeric protein is produced that converts the non-toxic prodrug 5-fluorocytosine (5-FC) into highly cytotoxic 5-fluorouracil (5-FU) and directly into 5-fluorouridine monophosphate (5-FUMP) thereby bypassing an important mechanism of chemoresistance to 5-FU. RESULTS: MeV-SCD was demonstrated to infect, replicate in and effectively lyse not only human ovarian cancer cell lines, but also primary tumor cells (albeit at lower efficiencies) that were derived from malignant ascites of ovarian cancer patients. Addition of the prodrug 5-FC significantly enhanced cell killing. Importantly, precision-cut tumor slices of human ovarian cancer patient specimens were efficiently infected with MeV-SCD. The prodrug-converting enzyme SCD was expressed by all infected tumor slices, thereby ensuring provision of the suicide gene arming function in patient-derived materials. CONCLUSIONS: With respect to safety and therapeutic impact, arming of oncolytic measles vaccine virus warrants further clinical investigation for ovarian cancer treatment.


Asunto(s)
Citosina Desaminasa/genética , Terapia Genética , Virus del Sarampión/genética , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/terapia , Pentosiltransferasa/genética , Línea Celular Tumoral , Femenino , Flucitosina/farmacología , Humanos , Vacuna Antisarampión , Saccharomyces cerevisiae/enzimología
4.
Gene Ther ; 20(11): 1033-41, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23719065

RESUMEN

Due to late diagnosis and a pronounced chemoresistance, most patients with hepatocellular carcinoma (HCC) have an overall poor prognosis. Measles vaccine viruses (MeV) have been shown to possess anti-tumor properties and their efficacy has been enhanced by arming with suicide genes. To test armed MeV for the treatment of HCC, we equipped it with the suicide gene Super-cytosine deaminase (SCD) and tested the efficacy in cell culture and in a mouse xenograft model of human HCC. Prodrug conversion was investigated in cell culture and quantified by high-performance liquid chromatography. We observed a strong oncolytic activity of MeV-SCD against human HCC in vitro and in vivo. The prodrug was efficiently converted in infected cells leading to a significant enhancement of the cytotoxic effect. Treatment of HCC xenografts with MeV caused long-term virus replication in tumor tissue. We show that the suicide gene therapy induces an apoptosis-like cell death but is not dependent on intact apoptosis pathways. These results demonstrate that MeV-based suicide gene therapy is a promising novel therapy regimen for HCC overcoming resistance towards conventional therapy. The independence from apoptosis raises hopes for the treatment of patients whose tumor cells exert defects in this cell death mechanism.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular/terapia , Citosina Desaminasa/genética , Virus del Sarampión , Viroterapia Oncolítica , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Chlorocebus aethiops , Cromatografía Liquida , Terapia Combinada , Citosina Desaminasa/metabolismo , Resistencia a Antineoplásicos , Genes Transgénicos Suicidas , Terapia Genética , Células Hep G2 , Humanos , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/terapia , Vacuna Antisarampión , Virus del Sarampión/genética , Ratones , Ratones Desnudos , Virus Oncolíticos/genética , Células Tumorales Cultivadas , Células Vero , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Gene Ther ; 19(3): 181-91, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22076043

RESUMEN

First-line treatment of recurrent and/or refractory head and neck squamous cell carcinoma (HNSCC) is based on platinum, 5-fluorouracil (5-FU) and the monoclonal antiEGFR antibody cetuximab. However, in most cases this chemoimmunotherapy does not cure the disease, and more than 50% of HNSCC patients are dying because of local recurrence of the tumors. In the majority of cases, HNSCC overexpress the epidermal growth factor receptor (EGFR), and its presence is associated with a poor outcome. In this study, we engineered an EGFR-targeted oncolytic measles virus (MV), armed with the bifunctional enzyme cytosine deaminase/uracil phosphoribosyltransferase (CD/UPRT). CD/UPRT converts 5-fluorocytosine (5-FC) into the chemotherapeutic 5-FU, a mainstay of HNSCC chemotherapy. This virus efficiently replicates in and lyses primary HNSCC cells in vitro. Arming with CD/UPRT mediates efficient prodrug activation with high bystander killing of non-infected tumor cells. In mice bearing primary HNSCC xenografts, intratumoral administration of MV-antiEGFR resulted in statistically significant tumor growth delay and prolongation of survival. Importantly, combination with 5-FC is superior to virus-only treatment leading to significant tumor growth inhibition. Thus, chemovirotherapy with EGFR-targeted and CD/UPRT-armed MV is highly efficacious in preclinical settings with direct translational implications for a planned Phase I clinical trial of MV for locoregional treatment of HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Citosina Desaminasa/genética , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/terapia , Virus del Sarampión/fisiología , Viroterapia Oncolítica/métodos , Pentosiltransferasa/genética , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/virología , Línea Celular Tumoral , Chlorocebus aethiops , Citosina Desaminasa/biosíntesis , Citosina Desaminasa/metabolismo , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Femenino , Flucitosina/farmacocinética , Flucitosina/farmacología , Fluorouracilo/farmacocinética , Fluorouracilo/farmacología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/virología , Humanos , Virus del Sarampión/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pentosiltransferasa/biosíntesis , Pentosiltransferasa/metabolismo , Profármacos/farmacocinética , Carcinoma de Células Escamosas de Cabeza y Cuello , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Gene Ther ; 18(8): 598-608, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21701532

RESUMEN

No curative therapy is currently available for locally advanced or metastatic pancreatic cancer. Therefore, new therapeutic approaches must be considered. Measles virus (MV) vaccine strains have shown promising oncolytic activity against a variety of tumor entities. For specific therapy of pancreatic cancer, we generated a fully retargeted MV that enters cells exclusively through the prostate stem cell antigen (PSCA). Besides a high-membrane frequency on prostate cancer cells, this antigen is expressed on pancreatic adenocarcinoma, but not on non-neoplastic tissue. PSCA expression levels differ within heterogeneous tumor bulks and between human pancreatic cell lines, and we could show specific infection of pancreatic adenocarcinoma cell lines with both high- and low-level PSCA expression. Furthermore, we generated a fully retargeted and armed MV-PNP-anti-PSCA to express the prodrug convertase purine nucleoside phosphorylase (PNP). PNP, which activates the prodrug fludarabine effectively, enhanced the oncolytic efficacy of the virus on infected and bystander cells. Beneficial therapeutic effects were shown in a pancreatic cancer xenograft model. Moreover, in the treatment of gemcitabine-resistant pancreatic adenocarcinoma cells, no cross-resistance to both MV oncolysis and activated prodrug was detected.


Asunto(s)
Adenocarcinoma/terapia , Virus del Sarampión/fisiología , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Adenocarcinoma/virología , Animales , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Chlorocebus aethiops , Terapia Combinada , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Masculino , Virus del Sarampión/inmunología , Virus del Sarampión/metabolismo , Ratones , Ratones SCID , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/virología , Profármacos/administración & dosificación , Profármacos/farmacocinética , Purina-Nucleósido Fosforilasa/genética , Purina-Nucleósido Fosforilasa/metabolismo , Células Vero , Vidarabina/administración & dosificación , Vidarabina/análogos & derivados , Vidarabina/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Gene Ther ; 11(7): 599-608, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14724676

RESUMEN

Current concerns over insertional mutagenesis by retroviral vectors mitigate investigations into alternative, potentially persistent gene therapy vector systems not dependent on genomic integration, such as Sendai virus vectors (SeVV). Prenatal gene therapy requires efficient gene delivery to several tissues, which may not be achievable by somatic gene transfer to the adult. Initially, to test the potential and tropism of the SeVV for gene delivery to fetal tissues, first-generation (replication- and propagation-competent) recombinant SeVV, expressing beta-galactosidase was introduced into late gestation immunocompetent mice via the amniotic and peritoneal cavities and the yolk sac vessels. At 2 days, this resulted in very high levels of expression particularly in the airway epithelium, mesothelium and vascular endothelium, respectively. However, as expected, substantial vector toxicity was observed. The efficiency of gene transfer and the level of gene expression were then examined using a second-generation SeVV. The second generation was developed to be still capable of cytoplasmic RNA replication and therefore high-level gene expression, but incapable of vector spread due to lack of the gene for viral F-protein. Vector was introduced into the fetal amniotic and peritoneal cavities, intravascularly, intramuscularly and intraspinally; at 2 days, expression was observed in the airway epithelia, peritoneal mesothelia, unidentified cells in the gut wall, locally at the site of muscle injection and in the dorsal root ganglia, respectively. Mortality was dramatically diminished compared with the first-generation vector.


Asunto(s)
Enfermedades Fetales/terapia , Feto/metabolismo , Terapia Genética/métodos , Vectores Genéticos/toxicidad , Enfermedades Pulmonares/terapia , Proteínas Virales de Fusión/genética , Adenoviridae/genética , Animales , Animales Recién Nacidos , Virus Defectuosos , Inyecciones Intravenosas , Pulmón/enzimología , Enfermedades Pulmonares/embriología , Ratones , Ratones Endogámicos , Modelos Animales , Virus Sendai/genética , beta-Galactosidasa/genética
8.
Biol Chem ; 379(11): 1359-65, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9865610

RESUMEN

The bifunctional protein DCoH (dimerization cofactor of HNF-1) is a transcriptional coactivator of homeodomain proteins and a cytoplasmic enzyme. Here we report on the isolation, nucleotide sequence and genomic structure of the chicken and rat DCoH genes. They have an unusual first exon coding for a single amino acid. The promoters are located within CpG-islands and lack TATA and CAAT consensus sequences. Analysis of the number of CpG dinucleotides in the promoter indicates little changes since man and chicken were separated in evolution whereas a 67% reduction was observed in the rat lineage. The DNA sequences were obtained entirely without cloning. For walking into unknown genomic regions we describe a modification of the ligation-mediated polymerase chain reaction (LM-PCR) that can replace conventional screening and cloning methods and greatly expedite the isolation of genomic DNA.


Asunto(s)
Hidroliasas/genética , Reacción en Cadena de la Polimerasa/métodos , Animales , Secuencia de Bases , Pollos , ADN Complementario , Regiones Promotoras Genéticas , Ratas , Mapeo Restrictivo , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...