Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biometals ; 32(3): 545-561, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31209680

RESUMEN

Oncolytic viruses rewire the immune system and can lead to long-lasting antitumor defenses against primary and metastatic tumors. However, results from clinical studies have shown heterogeneity in responses suggesting that multiplexed approaches may be necessary to consistently generate positive outcomes in patients. To this end, we explored the combination of oncolytic rhabdovirus VSV∆51 with vanadium(V) dipicolinate derivatives, which have already been explored for their antidiabetic properties in animal models. The combination of vanadium-based dipicolinate compounds with VSV∆51 significantly increased viral replication and cytotoxicity in the human renal cell carcinoma cell line 786-0. The effects of three vanadium(V)-dipicolinate coordination complexes ([VO2dipic]-, [VO2dipic-OH]- and [VO2dipic-Cl]- with -OH or -Cl in the para position) were compared to that of the simple salts using spectroscopy and speciation profiles. Like the vanadate salts and the vanadyl cation, all dioxovanadium(V) dipicolinate complexes tested were found to increase viral infection and cytotoxicity when used in combination with VSV∆51. Viral sensitization is dependent on the vanadium since free dipicolinate ligands exerted no effect on viral infection and viability. The ability of these complexes to interact with interfaces and the stability of the complexes were evaluated under physiological conditions. Results indicate that these complexes undergo hydrolysis in cell culture media thereby generating vanadate. The vanadium dipicolinate derivatives in the context of immunovirotherapy shares similarities with previous studies exploring the antidiabetic properties of the compounds. The synergy between vanadium compounds and the oncolytic virus suggests that these compounds may be valuable in the development of novel and effective pharmaco-viral therapies.


Asunto(s)
Antivirales/farmacología , Complejos de Coordinación/farmacología , Viroterapia Oncolítica , Virus Oncolíticos/efectos de los fármacos , Ácidos Picolínicos/farmacología , Compuestos de Vanadio/farmacología , Virosis/terapia , Antivirales/síntesis química , Antivirales/química , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Relación Dosis-Respuesta a Droga , Humanos , Concentración de Iones de Hidrógeno , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Ácidos Picolínicos/química , Relación Estructura-Actividad , Células Tumorales Cultivadas , Compuestos de Vanadio/química , Virosis/tratamiento farmacológico
2.
Cell Mol Life Sci ; 74(9): 1681-1692, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28004147

RESUMEN

Biotherapeutics have revolutionized modern medicine by providing medicines that would not have been possible with small molecules. With respect to cancer therapies, this represents the current sector of the pharmaceutical industry having the largest therapeutic impact, as exemplified by the development of recombinant antibodies and cell-based therapies. In cancer, one of the most common regulatory alterations is the perturbation of translational control. Among these, changes in eukaryotic initiation factor 4F (eIF4F) are associated with tumor initiation, progression, and drug resistance in a number of settings. This, coupled with the fact that systemic suppression of eIF4F appears well tolerated, indicates that therapeutic agents targeting eIF4F hold much therapeutic potential. Here, we discuss opportunities offered by biologicals for this purpose.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Factor 4F Eucariótico de Iniciación/antagonistas & inhibidores , Animales , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Factor 4F Eucariótico de Iniciación/metabolismo , Humanos , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos
3.
Mar Drugs ; 16(6)2018 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880736

RESUMEN

Lectins play diverse roles in physiological processes as biological recognition molecules. In this report, a gene encoding Tachypleus tridentatus Lectin (TTL) was inserted into an oncolytic vaccinia virus (oncoVV) vector to form oncoVV-TTL, which showed significant antitumor activity in a hepatocellular carcinoma mouse model. Furthermore, TTL enhanced oncoVV replication through suppressing antiviral factors expression such as interferon-inducible protein 16 (IFI16), mitochondrial antiviral signaling protein (MAVS) and interferon-beta (IFN-ß). Further investigations revealed that oncoVV-TTL replication was highly dependent on ERK activity. This study might provide insights into a novel way of the utilization of TTL in oncolytic viral therapies.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Cangrejos Herradura/metabolismo , Lectinas/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Virus Oncolíticos/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Antivirales/farmacología , Carcinoma Hepatocelular/virología , Línea Celular , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , Células HEK293 , Humanos , Interferón beta/metabolismo , Neoplasias Hepáticas/virología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Viroterapia Oncolítica/métodos , Fosfoproteínas/metabolismo , Virus Vaccinia/efectos de los fármacos
4.
Gene Ther ; 22(12): 947-59, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26196249

RESUMEN

Oncolytic viruses (OV) have broad potential as an adjuvant for the treatment of solid tumors. The present study addresses the feasibility of clinically applicable drugs to enhance the oncolytic potential of the OV Delta24-RGD in glioblastoma. In total, 446 drugs were screened for their viral sensitizing properties in glioblastoma stem-like cells (GSCs) in vitro. Validation was done for 10 drugs to determine synergy based on the Chou Talalay assay. Mechanistic studies were undertaken to assess viability, replication efficacy, viral infection enhancement and cell death pathway induction in a selected panel of drugs. Four viral sensitizers (fluphenazine, indirubin, lofepramine and ranolazine) were demonstrated to reproducibly synergize with Delta24-RGD in multiple assays. After validation, we underscored general applicability by testing candidate drugs in a broader context of a panel of different GSCs, various solid tumor models and multiple OVs. Overall, this study identified four viral sensitizers, which synergize with Delta24-RGD and two other strains of OVs. The viral sensitizers interact with infection, replication and cell death pathways to enhance efficacy of the OV.


Asunto(s)
Glioblastoma/terapia , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Flufenazina/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/virología , Células HCT116 , Humanos , Indoles/farmacología , Virus Oncolíticos/fisiología , Replicación Viral/efectos de los fármacos
5.
J Virol ; 88(5): 2927-40, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371063

RESUMEN

Vesicular stomatitis virus (VSV) is an oncolytic virus that induces cancer cell death through activation of the apoptotic pathway. Intrinsic resistance to oncolysis is found in some cell lines and many primary tumors as a consequence of residual innate immunity to VSV. In resistant-tumor models, VSV oncolytic potential can be reversibly stimulated by combination with epigenetic modulators, such as the histone deacetylase inhibitor vorinostat. Based on this reversible effect of vorinostat, we reasoned that critical host genes involved in oncolysis may likewise be reversibly regulated by vorinostat. A transcriptome analysis in prostate cancer PC3 cells identified a subset of NF-κB target genes reversibly regulated by vorinostat, as well as a group of interferon (IFN)-stimulated genes (ISGs). Consistent with the induction of NF-κB target genes, vorinostat-mediated enhancement of VSV oncolysis increased hyperacetylation of NF-κB RELA/p65. Additional bioinformatics analysis revealed that NF-κB signaling also increased the expression of several autophagy-related genes. Kinetically, autophagy preceded apoptosis, and apoptosis was observed only when cells were treated with both VSV and vorinostat. VSV replication and cell killing were suppressed when NF-κB signaling was inhibited using pharmacological or genetic approaches. Inhibition of autophagy by 3-methyladenine (3-MA) enhanced expression of ISGs, and either 3-MA treatment or genetic ablation of the autophagic marker Atg5 decreased VSV replication and oncolysis. Together, these data demonstrate that vorinostat stimulates NF-κB activity in a reversible manner via modulation of RELA/p65 signaling, leading to induction of autophagy, suppression of the IFN-mediated response, and subsequent enhancement of VSV replication and apoptosis.


Asunto(s)
Autofagia , Inhibidores de Histona Desacetilasas/farmacología , FN-kappa B/metabolismo , Virus Oncolíticos/efectos de los fármacos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Acetilación , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Cromatina/metabolismo , Análisis por Conglomerados , Técnicas de Silenciamiento del Gen , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , FN-kappa B/antagonistas & inhibidores , Viroterapia Oncolítica , Virus Oncolíticos/genética , Neoplasias de la Próstata/terapia , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transcriptoma , Virus de la Estomatitis Vesicular Indiana/genética , Replicación Viral , Vorinostat
6.
J Transl Med ; 13: 216, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26149494

RESUMEN

BACKGROUND: Oncolytic virotherapy is a novel approach for the treatment of glioblastoma multiforme (GBM) which is still a fatal disease. Pathologic features of GBM are characterized by the infiltration with microglia/macrophages and a strong interaction between immune- and glioma cells. The aim of this study was to determine the role of microglia and astrocytes for oncolytic vaccinia virus (VACV) therapy of GBM. METHODS: VACV LIVP 1.1.1 replication in C57BL/6 and Foxn1(nu/nu) mice with and without GL261 gliomas was analyzed. Furthermore, immunohistochemical analysis of microglia and astrocytes was investigated in non-, mock-, and LIVP 1.1.1-infected orthotopic GL261 gliomas in C57BL/6 mice. In cell culture studies virus replication and virus-mediated cell death of GL261 glioma cells was examined, as well as in BV-2 microglia and IMA2.1 astrocytes with M1 or M2 phenotypes. Co-culture experiments between BV-2 and GL261 cells and apoptosis/necrosis studies were performed. Organotypic slice cultures with implanted GL261 tumor spheres were used as additional cell culture system. RESULTS: We discovered that orthotopic GL261 gliomas upon intracranial virus delivery did not support replication of LIVP 1.1.1, similar to VACV-infected brains without gliomas. In addition, recruitment of Iba1(+) microglia and GFAP(+) astrocytes to orthotopically implanted GL261 glioma sites occurred already without virus injection. GL261 cells in culture showed high virus replication, while replication in BV-2 and IMA2.1 cells was barely detectable. The reduced viral replication in BV-2 cells might be due to rapid VACV-induced apoptotic cell death. In BV-2 and IMA 2.1 cells with M1 phenotype a further reduction of virus progeny and virus-mediated cell death was detected. Application of BV-2 microglial cells with M1 phenotype onto organotypic slice cultures with implanted GL261 gliomas resulted in reduced infection of BV-2 cells, whereas GL261 cells were well infected. CONCLUSION: Our results indicate that microglia and astrocytes, dependent on their activation state, may preferentially clear viral particles by immediate uptake after delivery. By acting as VACV traps they further reduce efficient virus infection of the tumor cells. These findings demonstrate that glia cells need to be taken into account for successful GBM therapy development.


Asunto(s)
Astrocitos/patología , Glioma/patología , Glioma/virología , Microglía/patología , Virus Oncolíticos/fisiología , Virus Vaccinia/fisiología , Replicación Viral , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/virología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Citometría de Flujo , Humanos , Inyecciones Intralesiones , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Necrosis , Virus Oncolíticos/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Virus Vaccinia/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
Gene Ther ; 21(4): 371-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24553345

RESUMEN

Oncolytic virotherapy is a promising strategy for reducing tumor burden through selective virus replication in rapidly proliferating cells. However, the lysis of slowly replicating cancer stem cells (CSCs), which maintain neoplastic clonality, is relatively modest and the potential contribution of programmed cell death pathways to oncolytic activity is still poorly understood. We show that the oncolytic virus ΔPK lyses CSC-enriched breast cancer and melanoma 3D spheroid cultures at low titers (0.1 pfu/cell) without resistance development and it inhibits the 3D growth potential (spheroids and agarose colonies) of melanoma and breast cancer cells. ΔPK induces calpain activation in both melanoma and breast cancer 3D cultures as determined by the loss of the p28 regulatory subunit, and 3D growth is restored by treatment with the calpain inhibitor PD150606. In melanoma, ΔPK infection also induces light chain 3 (LC3)-II accumulation and p62/SQSTM1 clearance, both markers of autophagy, and 3D growth is restored by treatment with the autophagy inhibitor chloroquine (CQ). However, expression of the autophagy-required protein Atg5 is not altered and CQ does not restore p62/SQSTM1 expression, suggesting that the CQ effect may be autophagy-independent. PD150606 restores expression of p62/SQSTM1 in ΔPK-infected melanoma cultures, suggesting that calpain activation induces anti-tumor activity through p62/SQSTM1 clearance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Melanoma/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Acrilatos/administración & dosificación , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Calpaína/administración & dosificación , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Melanoma/terapia , Melanoma/virología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/virología , Virus Oncolíticos/efectos de los fármacos , Proteína Sequestosoma-1 , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
8.
BMC Cancer ; 14: 551, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25078870

RESUMEN

BACKGROUND: Oncolytic viruses represent a promising therapy against cancers with acquired drug resistance. However, low efficacy limits its clinical application. The objective of this study is to investigate whether pharmacologically modulating autophagy could enhance oncolytic Newcastle disease virus (NDV) strain NDV/FMW virotherapy of drug-resistant lung cancer cells. METHODS: The effect of NDV/FMW infection on autophagy machinery in A549 lung cancer cell lines resistant to cisplatin (A549/DDP) or paclitaxel (A549/PTX) was investigated by detection of GFP-microtubule-associated protein 1 light chain 3 (GFP-LC3) puncta, formation of double-membrane vesicles and conversion of the nonlipidated form of LC3 (LC3-I) to the phosphatidylethanolamine-conjugated form (LC3-II). The effects of autophagy inhibitor chloroquine (CQ) and autophagy inducer rapamycin on NDV/FMW-mediated antitumor activity were evaluated both in culture cells and in mice bearing drug-resistant lung cancer cells. RESULTS: We show that NDV/FMW triggers autophagy in A549/PTX cells via dampening the class I PI3K/Akt/mTOR/p70S6K pathway, which inhibits autophagy. On the contrary, NDV/FMW infection attenuates the autophagic process in A549/DDP cells through the activation of the negative regulatory pathway. Furthermore, combination with CQ or knockdown of ATG5 significantly enhances NDV/FMW-mediated antitumor effects on A549/DDP cells, while the oncolytic efficacy of NDV/FMW in A549/PTX cells is significantly improved by rapamycin. Interestingly, autophagy modulation does not increase virus progeny in these drug resistant cells. Importantly, CQ or rapamycin significantly potentiates NDV/FMW oncolytic activity in mice bearing A549/DDP or A549/PTX cells respectively. CONCLUSIONS: These results demonstrate that combination treatment with autophagy modulators is an effective strategy to augment the therapeutic activity of NDV/FMW against drug-resistant lung cancers.


Asunto(s)
Autofagia/efectos de los fármacos , Cloroquina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/terapia , Virus de la Enfermedad de Newcastle/efectos de los fármacos , Viroterapia Oncolítica , Sirolimus/farmacología , Animales , Línea Celular Tumoral , Embrión de Pollo , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/genética , Trasplante de Neoplasias , Viroterapia Oncolítica/métodos , Virus Oncolíticos/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Ther ; 21(11): 2063-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23883863

RESUMEN

Current research has evaluated the intrinsic tumor-tropic properties of stem cell carriers for targeted anticancer therapy. Our laboratory has been extensively studying in the preclinical setting, the role of neural stem cells (NSCs) as delivery vehicles of CRAd-S-pk7, a gliomatropic oncolytic adenovirus (OV). However, the mediated toxicity of therapeutic payloads, such as oncolytic adenoviruses, toward cell carriers has significantly limited this targeted delivery approach. Following this rationale, in this study, we assessed the role of a novel antioxidant thiol, N-acetylcysteine amide (NACA), to prevent OV-mediated toxicity toward NSC carriers in an orthotropic glioma xenograft mouse model. Our results show that the combination of NACA and CRAd-S-pk7 not only increases the viability of these cell carriers by preventing reactive oxygen species (ROS)-induced apoptosis of NSCs, but also improves the production of viral progeny in HB1.F3.CD NSCs. In an intracranial xenograft mouse model, the combination treatment of NACA and NSCs loaded with CRAd-S-pk7 showed enhanced CRAd-S-pk7 production and distribution in malignant tissues, which improves the therapeutic efficacy of NSC-based targeted antiglioma oncolytic virotherapy. These data demonstrate that the combination of NACA and NSCs loaded with CRAd-S-pk7 may be a desirable strategy to improve the therapeutic efficacy of antiglioma oncolytic virotherapy.


Asunto(s)
Acetilcisteína/análogos & derivados , Adenoviridae/efectos de los fármacos , Glioma/terapia , Células-Madre Neurales/trasplante , Viroterapia Oncolítica/métodos , Acetilcisteína/farmacología , Adenoviridae/genética , Animales , Línea Celular Tumoral , Glioma/patología , Ratones , Ratones Desnudos , Terapia Molecular Dirigida/métodos , Neoplasias Experimentales , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Gene Med ; 15(3-4): 134-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23606319

RESUMEN

BACKGROUND: The oncolytic adenovirus Delta24-RGD is currently being tested in phase I trials for the treatment of glioblastoma (GBM). Literature suggests that frequently prescribed anticonvulsants for these patients, phenytoin (PHE), valproic acid (VPA) and levetiracetam (LEV), may interfere with cellular mechanisms of cancer or oncolytic virus activity. We therefore investigated the direct effects of these drugs on Delta24-RGD infection and oncolytic activity. METHODS: The anticonvulsants PHE, VPA, and LEV were combined with Delta24-RGD treatment in established glioma cell lines as well as on a panel of patient-derived GBM cultures. Effects on infection efficiency were assessed using luciferase-encoding adenoviral vectors. Oncolytic activity was determined by WST-1 assay and viral progeny production was quantified by dilution titration. RESULTS: IC50 values of the anti-epileptic drugs on the four glioma cell lines were far above clinically-relevant concentrations. At therapeutic concentrations, the anti-epileptics generally did not alter the infection efficiency of RGD-modified adenovirus, nor affect progeny production or oncolytic activity of Delta24-RGD. The only exception was found in U373 cells, where VPA slightly antagonised the oncolytic effect of Delta24-RGD (from 29% to 55% viability, p<0.01) as well as viral progeny production (60% decrease, p<0.01). Oncolysis by Delta24-RGD was not inhibited by the anti-epileptics in any of the patient-derived glioma cultures (n=6). In fact, in one culture a slight enhancement of viral oncolysis by PHE and LEV was found, from 89.7% viability to 76% and 62.4%, respectively (p<0.01) CONCLUSIONS: Therapeutic levels of valproic acid, phenytoin and levetiracetam do not negatively interfere with the infection efficiency or oncolytic activity of Delta24-RGD in patient-derived GBM cells. Therefore, there is no indication that the choice of anticonvulsant for seizure control in glioma patients should take treatment with Delta24-RGD into account.


Asunto(s)
Adenoviridae/efectos de los fármacos , Anticonvulsivantes/farmacología , Glioma/tratamiento farmacológico , Viroterapia Oncolítica/métodos , Virus Oncolíticos/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Levetiracetam , Luciferasas , Fenitoína , Piracetam/análogos & derivados , Ácido Valproico
11.
J Transl Med ; 11: 257, 2013 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-24124726

RESUMEN

BACKGROUND: Gene therapy using a recombinant adenovirus (Ad) encoding secretory human endostatin (Ad-Endo) has been demonstrated to be a promising antiangiogenesis and antitumor strategy of in animal models and clinical trials. The E1B55KD-deficient Ad dl1520 was also found to replicate selectively in and destroy cancer cells. In this study, we aimed to investigate the antitumor effects of antiangiogenic agent Ad-Endo combined with the oncolytic Ad dl1520 on gastric cancer (GC) in vitro and in vivo and determine the mechanisms of these effects. METHODS: The Ad DNA copy number was determined by real-time PCR, and gene expression was assessed by ELISA, Western blotting or immunohistochemistry. The anti-proliferation effect (cytotoxicity) of Ad was assessed using the colorimetry-based MTT cell viability assay. The antitumor effects were evaluated in BALB/c nude mice carrying SGC-7901 GC xenografts. The microvessel density and Ad replication in tumor tissue were evaluated by checking the expression of CD34 and hexon proteins, respectively. RESULTS: dl1520 replicated selectively in GC cells harboring an abnormal p53 pathway, including p53 mutation and the loss of p14(ARF) expression, but did not in normal epithelial cells. In cultured GC cells, dl1520 rescued Ad-Endo replication, and dramatically promoted endostatin expression by Ad-Endo in a dose- and time-dependent manner. In turn, the addition of Ad-Endo enhanced the inhibitory effect of dl1520 on the proliferation of GC cells. The transgenic expression of Ad5 E1A and E1B19K simulated the rescue effect of dl1520 supporting Ad-Endo replication in GC cells. In the nude mouse xenograft model, the combined treatment with dl1520 and Ad-Endo significantly inhibited tumor angiogenesis and the growth of GC xenografts through the increased endostatin expression and oncolytic effects. CONCLUSIONS: Ad-Endo combined with dl1520 has more antitumor efficacy against GC than Ad-Endo or dl1520 alone. These findings indicate that the combination of Ad-mediated antiangiogenic gene therapy and oncolytic Ad therapeutics could be one of promising comprehensive treatment strategies for GC.


Asunto(s)
Adenoviridae/metabolismo , Antineoplásicos/uso terapéutico , Endostatinas/uso terapéutico , Recombinación Genética/genética , Neoplasias Gástricas/tratamiento farmacológico , Proteínas Virales/metabolismo , Adenoviridae/efectos de los fármacos , Proteínas E1B de Adenovirus/metabolismo , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Endostatinas/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/patología , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Transl Med ; 11: 216, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-24044901

RESUMEN

BACKGROUND: The brain cancer stem cell (CSC) model describes a small subset of glioma cells as being responsible for tumor initiation, conferring therapy resistance and tumor recurrence. In brain CSC, the PI3-K/AKT and the RAS/mitogen activated protein kinase (MAPK) pathways are found to be activated. In consequence, the human transcription factor YB-1, knowing to be responsible for the emergence of drug resistance and driving adenoviral replication, is phosphorylated and activated. With this knowledge, YB-1 was established in the past as a biomarker for disease progression and prognosis. This study determines the expression of YB-1 in glioblastoma (GBM) specimen in vivo and in brain CSC lines. In addition, the capacity of Ad-Delo3-RGD, an YB-1 dependent oncolytic adenovirus, to eradicate CSC was evaluated both in vitro and in vivo. METHODS: YB-1 expression was investigated by immunoblot and immuno-histochemistry. In vitro, viral replication as well as the capacity of Ad-Delo3-RGD to replicate in and, in consequence, to kill CSC was determined by real-time PCR and clonogenic dilution assays. In vivo, Ad-Delo3-RGD-mediated tumor growth inhibition was evaluated in an orthotopic mouse GBM model. Safety and specificity of Ad-Delo3-RGD were investigated in immortalized human astrocytes and by siRNA-mediated downregulation of YB-1. RESULTS: YB-1 is highly expressed in brain CSC lines and in GBM specimen. Efficient viral replication in and virus-mediated lysis of CSC was observed in vitro. Experiments addressing safety aspects of Ad-Delo3-RGD showed that (i) virus production in human astrocytes was significantly reduced compared to wild type adenovirus (Ad-WT) and (ii) knockdown of YB-1 significantly reduced virus replication. Mice harboring othotopic GBM developed from a temozolomide (TMZ)-resistant GBM derived CSC line which was intratumorally injected with Ad-Delo3-RGD survived significantly longer than mice receiving PBS-injections or TMZ treatment. CONCLUSION: The results of this study supported YB-1 based virotherapy as an attractive therapeutic strategy for GBM treatment which will be exploited further in multimodal treatment concepts.


Asunto(s)
Adenoviridae/metabolismo , Neoplasias Encefálicas/patología , Glioma/patología , Células Madre Neoplásicas/patología , Virus Oncolíticos/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo , Adenoviridae/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glioma/enzimología , Humanos , Ratones , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/metabolismo , Virus Oncolíticos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Temozolomida , Proteínas Supresoras de Tumor/metabolismo , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Folia Biol (Praha) ; 59(2): 68-75, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23746172

RESUMEN

Oncolytic viruses infect, replicate in, and lyse tumour cells but spare the normal ones. One of oncolytic viruses is a naturally occurring replication-competent reovirus (RV), which preferentially kills tumour cells with activated Ras signaling pathways. The aim of this study was to survey effects of RV on brain tumour-derived cells in vitro under hypoxic conditions since hypoxia causes resistance to radio- and chemotherapy. This study demonstrates that RV replicates preferentially in tumour cells and that the virus is able to overcome cellular adaptation to hypoxia and infect and kill hypoxic tumour cells. RV can both replicate in hypoxic tumour microenvironment and cause the cytopathic effect, subsequently inducing cell death. We found that a large proportion of cells are killed in hypoxia (1% O2) by caspase-independent mechanisms. Furthermore, we learned that the cell death induced by RV in hypoxic conditions is not caused by autophagy.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Reoviridae/fisiología , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Caspasa 3/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibroblastos/virología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Virus Oncolíticos/efectos de los fármacos , Oxígeno/farmacología , Reoviridae/efectos de los fármacos , Infecciones por Reoviridae/patología , Infecciones por Reoviridae/virología
14.
Med Sci (Paris) ; 29(2): 165-73, 2013 Feb.
Artículo en Francés | MEDLINE | ID: mdl-23452603

RESUMEN

Since over a century, medical literature has reported cases of viral infections leading to tumour regression. This phenomenon, now understood, can be exploited for cancer therapy. It involves viruses defined as "oncolytic". These viruses, either wild-type or genetically engineered, replicate preferentially in malignant cells. They induce tumour regression through various mechanisms including direct cell lysis and stimulation of an anti-tumour immune response. Several oncolytic viruses have reached late-stage clinical investigation and could be approved soon for treating certain neoplasms. While already promising, there is still room for improvement and various genetic, immunological, and pharmacological strategies are currently under development to increase their therapeutic efficacy.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Animales , Genes Transgénicos Suicidas/genética , Genes Transgénicos Suicidas/fisiología , Vectores Genéticos/síntesis química , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Neoplasias/genética , Neoplasias/inmunología , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Terapias en Investigación
15.
Mol Cancer ; 11: 31, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22569271

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) has a dismal 5-year-survival rate of 10%, so novel strategies are warranted. IL-24 mediates anti-tumor activity reducing STAT3 expression, which suggests that interferon (IFN) alpha may augment tumor cell lysis and reduce angiogenesis. We investigated the antitumor activity of treatment with IFN-α, with the oncolytic adenovirus SG600-IL-24, or the combination of both in HCC in vitro and in vivo. RESULTS: RT-PCR, ELISA assay and Western-blot confirmed that the exogenous IL-24 gene was highly expressed in HCC cells infected with SG600-IL-24. Treatment with combined IFN-α and SG600-IL-24 suppressed growth and promoted apoptosis of the HepG2, MHCC97L, and HCCLM3 cell lines compared with the normal cell line L02. The combined therapy increased STAT1 and SOCS1 and apoptosis, but decreased the expression of the metastatic and angiogenic proteins MMP-2, XIAP, OPN, and VEGF, which are regulated by STAT3 in HCC cells in vitro. To assess the effects in vivo, the HCC cell line HCCLM3 was transplanted subcutaneously into the right flanks of nude mice. Mice in the IFN-α group, the SG600-IL-24 group, or the combined therapy group had significantly suppressed growth of the HCC xenografted tumors compared to the PBS control group of mice. Among the mice treated with the combination of IFN-α and SG600-IL-24, three of those eight mice had long-term survival and no evidence of a tumor. These mice also had decreased expression of the metastatic and angiogenic proteins MMP-2, XIAP, OPN, and VEGF. CONCLUSIONS: The present study demonstrated for the first time the potential antitumor activity of IFN-α combined with the oncolytic adenovirus SG600-IL-24 in HCC both in vitro and in vivo, and suggests its further development as a potential candidate for HCC cancer gene therapy.


Asunto(s)
Adenoviridae/metabolismo , Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Interferón-alfa/farmacología , Interleucinas/metabolismo , Neoplasias Hepáticas/metabolismo , Virus Oncolíticos/metabolismo , Adenoviridae/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Interleucinas/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Mol Ther ; 19(3): 490-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21081903

RESUMEN

Oncolytic adenoviruses show promise as a cancer treatment. However, they generate acute inflammatory responses with production of cytokines, including tumor necrosis factor-α (TNF-α). We investigated whether inhibition of TNF-α augments efficacy of the E1A CR2-deleted adenovirus dl922-947 in ovarian cancer. dl922-947 induced transcription of TNF-α and its downstream signaling targets interleukin-6 and -8 (IL-6 and IL-8) in ovarian cancer cells. In vitro, RNAi-mediated knockdown of TNF-α reduced production of multiple inflammatory cytokines after infection and increased ovarian cancer cell sensitivity to virus cytotoxicity, as did treatment with the anti-TNF-α antibody infliximab. In vivo, stable knockdown of TNF-α in IGROV-1 xenografts increased the anticancer activity of dl922-947. In addition, inhibition of TNF-α using monoclonal antibodies also improved dl922-947 efficacy. This increased efficacy resulted from suppression of cellular inhibitor of apoptosis-1 and -2 (cIAP1 and cIAP2) transcription in malignant cells and a consequent increase in caspase-mediated apoptosis. These findings suggest that TNF-α acts as a survival factor in adenovirus-infected cells. Combining TNF-α inhibition with oncolytic adenoviruses could improve antitumor activity in clinical trials.


Asunto(s)
Adenoviridae , Proteínas Inhibidoras de la Apoptosis/metabolismo , Virus Oncolíticos , Neoplasias Ováricas , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Adenoviridae/efectos de los fármacos , Adenoviridae/inmunología , Adenoviridae/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Infliximab , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Viroterapia Oncolítica , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/inmunología , Virus Oncolíticos/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Neoplasias Ováricas/virología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/farmacología , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Transl Res ; 240: 64-86, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34757194

RESUMEN

Oncolytic virotherapy is a new and safe therapeutic strategy for cancer treatment. In our previous study, a new type of oncolytic herpes simplex virus type 2 (oHSV2) was constructed. Following the completion of a preclinical study, oHSV2 has now entered into clinical trials for the treatment of melanoma and other solid tumors (NCT03866525). Oncolytic viruses (OVs) are generally able to directly destroy tumor cells and stimulate the immune system to fight tumors. Natural killer (NK) cells are important components of the innate immune system and critical players against tumor cells. But the detailed interactions between oncolytic viruses and NK cells and these interaction effects on the antitumor immune response remain to be elucidated. In particular, the functions of activating surface receptors and checkpoint inhibitors on oHSV2-treated NK cells and tumor cells are still unknown. In this study, we found that UV-oHSV2 potently activates human peripheral blood mononuclear cells, leading to increased antitumor activity in vitro and in vivo. Further investigation indicated that UV-oHSV2-stimulated NK cells release IFN-γ via Toll-like receptor 2 (TLR2)/NF-κB signaling pathway and exert antitumor activity via TLR2. We found for the first time that the expression of a pair of checkpoint molecules, NKG2A (on NK cells) and HLA-E (on tumor cells), is upregulated by UV-oHSV2 stimulation. Anti-NKG2A and anti-HLA-E treatment could further enhance the antitumor effects of UV-oHSV2-stimulated NK92 cells in vitro and in vivo. As our oHSV2 clinical trial is ongoing, we expect that the combination therapy of oncolytic virus oHSV2 and anti-NKG2A/anti-HLA-E antibodies may have synergistic antitumor effects in our future clinical trials.


Asunto(s)
Herpesvirus Humano 2/efectos de la radiación , Inhibidores de Puntos de Control Inmunológico/farmacología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Virus Oncolíticos/efectos de la radiación , Rayos Ultravioleta , Inactivación de Virus/efectos de la radiación , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Herpesvirus Humano 2/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Virus Oncolíticos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/metabolismo , Inactivación de Virus/efectos de los fármacos , Antígenos HLA-E
18.
Biochem Biophys Res Commun ; 406(2): 262-7, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21316343

RESUMEN

Genetically engineered Sindbis viruses (SIN) are excellent oncolytic agents in preclinical models. Several human cancers have aberrant Akt signaling, and kinase inhibitors including rapamycin are currently tested in combination therapies with oncolytic viruses. Therefore, it was of interest to delineate possible cross-regulation between SIN replication and PI3K/Akt/mTOR signaling. Here, using HEK293T cells as host, we report the following key findings: (a) robust SIN replication occurs in the presence of mTOR specific inhibitors, rapamycin and torin1 or Ly294002--a PI3K inhibitor, suggesting a lack of requirement for PI3K/Akt/mTOR signaling; (b) suppression of phosphorylation of Akt, mTOR and its effectors S6, and 4E-BP1 occurs late during SIN infection: a viral function that may be beneficial in counteracting cellular drug resistance to kinase inhibitors; (c) Ly294002 and SIN act additively to suppress PI3K/Akt/mTOR pathway with little effect on virus release; and (d) SIN replication induces host translational shut off, phosphorylation of eIF2α and apoptosis. This first report on the potent inhibition of Akt/mTOR signaling by SIN replication, bolsters further studies on the development and evaluation of engineered SIN genotypes in vitro and in vivo for unique cytolytic functions.


Asunto(s)
Virus Oncolíticos/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Virus Sindbis/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Replicación Viral/fisiología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular , Cromonas/farmacología , Ingeniería Genética , Células HEK293 , Humanos , Morfolinas/farmacología , Naftiridinas/farmacología , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas/metabolismo , Virus Sindbis/efectos de los fármacos , Virus Sindbis/genética , Sirolimus/farmacología , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
19.
Mol Ther ; 18(5): 903-11, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20179683

RESUMEN

The therapeutic potential of oncolytic adenoviruses is limited by the rate of adenovirus release. Based on the observation that several viruses induce cell death and progeny release by disrupting intracellular calcium homeostasis, we hypothesized that the alteration in intracellular calcium concentration induced by verapamil could improve the rate of virus release and spread, eventually enhancing the antitumoral activity of oncolytic adenoviruses. Our results indicate that verapamil substantially enhanced the release of adenovirus from a variety of cell types resulting in an improved cell-to-cell spread and cytotoxicity. Furthermore, the combination of the systemic administration of an oncolytic adenovirus (ICOVIR-5) with verapamil in vivo greatly improved its antitumoral activity in two different tumor xenograft models without affecting the selectivity of this virus. Overall, our findings indicate that verapamil provides a new, safe, and versatile way to improve the antitumoral potency of oncolytic adenoviruses in the clinical setting.


Asunto(s)
Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Verapamilo/uso terapéutico , Adenoviridae/efectos de los fármacos , Adenoviridae/genética , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/terapia , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/genética , Liberación del Virus/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Proc Natl Acad Sci U S A ; 105(39): 14981-6, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18815361

RESUMEN

Intratumoral innate immunity can play a significant role in blocking the effective therapeutic spread of a number of oncolytic viruses (OVs). Histone deacetylase inhibitors (HDIs) are known to influence epigenetic modifications of chromatin and can blunt the cellular antiviral response. We reasoned that pretreatment of tumors with HDIs could enhance the replication and spread of OVs within malignancies. Here, we show that HDIs markedly enhance the spread of vesicular stomatitis virus (VSV) in a variety of cancer cells in vitro, in primary tumor tissue explants and in multiple animal models. This increased oncolytic activity correlated with a dampening of cellular IFN responses and augmentation of virus-induced apoptosis. These results illustrate the general utility of HDIs as chemical switches to regulate cellular innate antiviral responses and to provide controlled growth of therapeutic viruses within malignancies. HDIs could have a profoundly positive impact on the clinical implementation of OV therapeutics.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Inhibidores de Histona Desacetilasas , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/efectos de los fármacos , Animales , Benzamidas/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Interferones/administración & dosificación , Masculino , Ratones , Ratones Endogámicos , Neoplasias/tratamiento farmacológico , Neoplasias/virología , Virus Oncolíticos/inmunología , Virus Oncolíticos/fisiología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/virología , Piridinas/uso terapéutico , Vesiculovirus/efectos de los fármacos , Vesiculovirus/inmunología , Vesiculovirus/fisiología , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA