Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Ther Oncolytics ; 21: 340-355, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34141871

RESUMEN

Advanced pancreatic cancer is characterized by few treatment options and poor outcomes. Oncolytic virotherapy and chemotherapy involve complementary pharmacodynamics and could synergize to improve therapeutic efficacy. Likewise, multimodality treatment may cause additional toxicity, and new agents have to be safe. Balancing both aims, we generated an oncolytic measles virus for 5-fluorouracil-based chemovirotherapy of pancreatic cancer with enhanced tumor specificity through microRNA-regulated vector tropism. The resulting vector encodes a bacterial prodrug convertase, cytosine deaminase-uracil phosphoribosyl transferase, and carries synthetic miR-148a target sites in the viral F gene. Combination of the armed and targeted virus with 5-fluorocytosine, a prodrug of 5-fluorouracil, resulted in cytotoxicity toward both infected and bystander pancreatic cancer cells. In pancreatic cancer xenografts, a single intratumoral injection of the virus induced robust in vivo expression of prodrug convertase. Based on intratumoral transgene expression kinetics, we devised a chemovirotherapy regimen to assess treatment efficacy. Concerted multimodality treatment with intratumoral virus and systemic prodrug administration delayed tumor growth and prolonged survival of xenograft-bearing mice. Our results demonstrate that 5-fluorouracil-based chemovirotherapy with microRNA-sensitive measles virus is an effective strategy against pancreatic cancer at a favorable therapeutic index that warrants future clinical translation.

2.
Oncoimmunology ; 6(4): e1285992, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28507792

RESUMEN

Combination of oncolytic virotherapy with immunomodulators is emerging as a promising therapeutic strategy for numerous tumor entities. In this study, we developed measles Schwarz vaccine strain vectors encoding immunomodulators to support different phases in the establishment of antitumor immune responses. Therapeutic efficacy of the novel vectors was evaluated in the immunocompetent MC38cea tumor model. We identified vectors encoding an IL-12 fusion protein (MeVac FmIL-12) and an antibody against PD-L1 (MeVac anti-PD-L1), respectively, as the most effective. Treatment of established tumors with MeVac FmIL-12 achieved 90% complete remissions. Profiling of the tumor immune microenvironment revealed activation of a type 1 T helper cell-directed response, with MeVac FmIL-12 ensuring potent early natural killer and effector T cell activation as well as upregulation of the effector cytokines IFN-γ and TNF-α. CD8+ T cells were found to be essential for the therapeutic efficacy of MeVac FmIL-12. Results of this study present MeVac FmIL-12 as a novel approach for targeted IL-12 delivery and elucidate mechanisms of successful immunovirotherapy.

3.
Mol Ther ; 22(11): 1949-59, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25156126

RESUMEN

We hypothesized that the combination of oncolytic virotherapy with immune checkpoint modulators would reduce tumor burden by direct cell lysis and stimulate antitumor immunity. In this study, we have generated attenuated Measles virus (MV) vectors encoding antibodies against CTLA-4 and PD-L1 (MV-aCTLA-4 and MV-aPD-L1). We characterized the vectors in terms of growth kinetics, antibody expression, and cytotoxicity in vitro. Immunotherapeutic effects were assessed in a newly established, fully immunocompetent murine model of malignant melanoma, B16-CD20. Analyses of tumor-infiltrating lymphocytes and restimulation experiments indicated a favorable immune profile after MV-mediated checkpoint modulation. Therapeutic benefits in terms of delayed tumor progression and prolonged median overall survival were observed for animals treated with vectors encoding anti-CTLA-4 and anti-PD-L1, respectively. Combining systemic administration of antibodies with MV treatment also improved therapeutic outcome. In vivo oncolytic efficacy against human tumors was studied in melanoma xenografts. MV-aCTLA-4 and MV-aPD-L1 were equally efficient as parental MV in this model, with high rates of complete tumor remission (> 80%). Furthermore, we could demonstrate lysis of tumor cells and transgene expression in primary tissue from melanoma patients. The current results suggest rapid translation of combining immune checkpoint modulation with oncolytic viruses into clinical application.


Asunto(s)
Antígeno B7-H1/metabolismo , Antígeno CTLA-4/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/terapia , Virus Oncolíticos/inmunología , Animales , Vectores Genéticos/administración & dosificación , Virus del Sarampión/genética , Virus del Sarampión/inmunología , Virus del Sarampión/metabolismo , Melanoma Experimental/inmunología , Ratones , Viroterapia Oncolítica , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Resultado del Tratamiento
4.
Hum Gene Ther ; 24(7): 644-54, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23642239

RESUMEN

Oncolytic measles viruses (MV) derived from the live attenuated vaccine strain have been engineered for increased antitumor activity, and are currently under investigation in clinical phase 1 trials. Approaches with other viral vectors have shown that insertion of immunomodulatory transgenes enhances the therapeutic potency. In this study, we engineered MV for expression of the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF). For the first time, therapeutic efficacy and adaptive immune response in the context of MV oncolysis could be evaluated in the previously established immunocompetent murine colon adenocarcinoma model MC38cea. MC38cea cells express the human carcinoembryonic antigen (CEA), allowing for infection with retargeted MV. Intratumoral application of MV-GMCSF significantly delayed tumor progression and prolonged median overall survival compared with control virus-treated mice. Importantly, more than one-third of mice treated with MV-GMCSF showed complete tumor remission and rejected successive tumor reengraftment, demonstrating robust long-term protection. An enhanced cell-mediated tumor-specific immune response could be detected by lactate dehydrogenase assay and interferon-γ enzyme-linked immunospot assay. Furthermore, MV-GMCSF treatment correlated with increased abundance of tumor-infiltrating CD3(+) lymphocytes analyzed by quantitative microscopy of tumor sections. These findings underline the potential of oncolytic, GM-CSF-expressing MV as an effective therapeutic cancer vaccine actively recruiting adaptive immune responses for enhanced therapeutic impact and tumor elimination. Thus, the treatment benefit of this combined immunovirotherapy approach has direct implications for future clinical trials.


Asunto(s)
Adenocarcinoma/prevención & control , Neoplasias del Colon/prevención & control , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Virus del Sarampión/inmunología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Inmunidad Adaptativa/inmunología , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Análisis de Varianza , Animales , Chlorocebus aethiops , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Ensayo de Immunospot Ligado a Enzimas , Ingeniería Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunohistoquímica , L-Lactato Deshidrogenasa/metabolismo , Virus del Sarampión/genética , Ratones , Virus Oncolíticos/genética , Células Vero
5.
J Invest Dermatol ; 133(4): 1034-42, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23223133

RESUMEN

Effective treatment modalities for advanced melanoma are desperately needed. An innovative approach is virotherapy, in which viruses are engineered to infect cancer cells, resulting in tumor cell lysis and an amplification effect by viral replication and spread. Ideally, tumor selectivity of these oncolytic viruses is already determined during viral cell binding and entry, which has not been reported for melanoma. We engineered an oncolytic measles virus entering melanoma cells through the high molecular weight melanoma-associated antigen (HMWMAA) and proved highly specific infection and spread in melanoma cells. We further enhanced this oncolytic virus by inserting the FCU1 gene encoding the yeast-derived prodrug convertases cytosine deaminase and uracil phosphoribosyltransferase. Combination treatment with armed and retargeted MV-FCU1-αHMWMAA and the prodrug 5-fluorocytosine (5-FC) led to effective prodrug conversion to 5-fluorouracil, extensive cytotoxicity to melanoma cells, and excessive bystander killing of noninfected cells. Importantly, HMWMAA-retargeted MV showed antitumor activity in a human xenograft mouse model, which was further increased by the FCU1/5-FC prodrug activation system. Finally, we demonstrated susceptibility of melanoma skin metastasis biopsies to HMWMAA-retargeted MV. The highly selective, entry-targeted and armed oncolytic virus MV-FCU1-αHMWMAA may become a potent building block of future melanoma therapies.


Asunto(s)
Virus del Sarampión/genética , Melanoma/tratamiento farmacológico , Viroterapia Oncolítica/métodos , Profármacos/farmacocinética , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antígenos de Neoplasias/metabolismo , Antimetabolitos/metabolismo , Biopsia , Línea Celular Tumoral , Chlorocebus aethiops , Terapia Combinada , Femenino , Flucitosina/metabolismo , Ingeniería Genética/métodos , Genoma Viral/genética , Humanos , Melanoma/secundario , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes/genética , Neoplasias Cutáneas/patología , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Ther ; 19(6): 1097-106, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21468006

RESUMEN

Oncolytic measles viruses (MV) derived from the live attenuated vaccine strain have been engineered for increased tumor-cell specificity, and are currently under investigation in clinical trials including a phase I study for glioblastoma multiforme (GBM). Recent preclinical studies have shown that the cellular tropism of several viruses can be controlled by inserting microRNA-target sequences into their genomes, thereby inhibiting spread in tissues expressing cognate microRNAs. Since neuron-specific microRNA-7 is downregulated in gliomas but highly expressed in normal brain tissue, we engineered a microRNA-sensitive virus containing target sites for microRNA-7 in the 3'-untranslated region of the viral fusion gene. In presence of microRNA-7 this modification inhibits translation of envelope proteins, restricts viral spread, and progeny production. Even though highly attenuated in presence of microRNA-7, this virus retained full efficacy against glioblastoma xenografts. Furthermore, microRNA-mediated inhibition protected genetically modified mice susceptible to MV infection from a potentially lethal intracerebral challenge. Importantly, endogenous microRNA-7 expression in primary human brain resections tightly restricted replication and spread of microRNA-sensitive virus. This is proof-of-concept that tropism restriction by tissue-specific microRNAs can be adapted to oncolytic MV to regulate viral replication and gene expression to maximize tumor specificity without compromising oncolytic efficacy.


Asunto(s)
Supervivencia Celular/fisiología , Virus del Sarampión/genética , MicroARNs/genética , Virus Oncolíticos/fisiología , Animales , Neoplasias Encefálicas/terapia , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Chlorocebus aethiops , Femenino , Vectores Genéticos/genética , Glioblastoma/terapia , Glioma/terapia , Humanos , Immunoblotting , Técnicas In Vitro , Virus del Sarampión/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Viroterapia Oncolítica , Virus Oncolíticos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Tissue Eng Part A ; 16(12): 3687-95, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20673025

RESUMEN

Biocements are clinically applied materials for bone replacement in non-load-bearing defects. Depending on their final composition, cements can be either resorbed or remain stable at the implantation site. Degradation can occur by two different mechanisms, by simple dissolution (passive) or after osteoclastic bone remodeling (active). This study investigated both the passive and active in vitro resorption behavior of brushite (CaHPO4 · 2H2O), monetite (CaHPO4), calcium-deficient hydroxyapatite (CDHA; Ca9(PO4)5HPO4OH), and struvite (MgNH4PO4 · 6H2O) cements. Passive resorption was measured by incubating the cement samples in a cell culture medium, whereas active resorption was determined during the surface culture of multinuclear osteoclastic cells derived from RAW 264.7 macrophages. Osteoclast formation was confirmed by showing tartrate resistant acid phosphatase (TRAP) activity on CDHA, brushite, and monetite surfaces, as well as by measuring calcitonin receptor (CT-R) expression as an osteoclast-specific protein by Western blot analysis for struvite ceramics. An absence of passive degradation and only marginally active degradation of <0.01% were found for CDHA matrices. For the secondary calcium phosphates brushite and monetite, active degradation was predominant with a cumulative Ca²+ release of 2.02 (1.20) µmol during 13 days, whereas passive degradation released only 0.788 (0.04) µmol calcium ions into the medium. The struvite cement was the most degradable with a passive (active) release of 9.26 (2.92) Mg²+ ions and a total weight loss of 4.7% over 13 days of the study.


Asunto(s)
Fosfatos de Calcio/metabolismo , Compuestos de Magnesio/metabolismo , Osteoclastos/metabolismo , Fosfatos/metabolismo , Animales , Western Blotting , Línea Celular , Espectrometría de Masas , Ratones , Microscopía Fluorescente , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA