Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Cell Death Dis ; 12(7): 663, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34230456

RESUMEN

A majority of mesothelioma specimens were defective of p14 and p16 expression due to deletion of the INK4A/ARF region, and the p53 pathway was consequently inactivated by elevated MDM2 functions which facilitated p53 degradaton. We investigated a role of p53 elevation by MDM2 inhibitors, nutlin-3a and RG7112, in cytotoxicity of replication-competent adenoviruses (Ad) lacking the p53-binding E1B55kDa gene (Ad-delE1B). We found that a growth inhibition by p53-activating Ad-delE1B was irrelevant to p53 expression in the infected cells, but combination of Ad-delE1B and the MDM2 inhibitor produced synergistic inhibitory effects on mesothelioma with the wild-type but not mutated p53 genotype. The combination augmented p53 phosphorylation, activated apoptotic but not autophagic pathway, and enhanced DNA damage signals through ATM-Chk2 phosphorylation. The MDM2 inhibitors facilitated production of the Ad progenies through augmented expression of nuclear factor I (NFI), one of the transcriptional factors involved in Ad replications. Knocking down of p53 with siRNA did not increase the progeny production or the NFI expression. We also demonstrated anti-tumor effects by the combination of Ad-delE1B and the MDM2 inhibitors in an orthotopic animal model. These data collectively indicated that upregulation of wild-type p53 expression contributed to cytotoxicity by E1B55kDa-defective replicative Ad through NFI induction and suggested that replication-competent Ad together with augmented p53 levels was a therapeutic strategy for p53 wild-type mesothelioma.


Asunto(s)
Adenoviridae/genética , Proteínas E1 de Adenovirus/genética , Antineoplásicos/farmacología , Imidazoles/farmacología , Imidazolinas/farmacología , Mesotelioma/terapia , Neurofibromina 1/metabolismo , Viroterapia Oncolítica , Virus Oncolíticos/genética , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Adenoviridae/crecimiento & desarrollo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Quimioterapia Adyuvante , Regulación Neoplásica de la Expresión Génica , Mesotelioma/genética , Mesotelioma/metabolismo , Mesotelioma/virología , Ratones Endogámicos BALB C , Ratones Desnudos , Neurofibromina 1/genética , Virus Oncolíticos/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mar Drugs ; 19(6)2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34064193

RESUMEN

Oncolytic vaccina virus (oncoVV) used for cancer therapy has progressed in recent years. Here, a gene encoding white-spotted charr lectin (WCL) was inserted into an oncoVV vector to form an oncoVV-WCL recombinant virus. OncoVV-WCL induced higher levels of apoptosis and cytotoxicity, and replicated faster than control virus in cancer cells. OncoVV-WCL promoted IRF-3 transcriptional activity to induce higher levels of type I interferons (IFNs) and blocked the IFN-induced antiviral response by inhibiting the activity of IFN-stimulated responsive element (ISRE) and the expression of interferon-stimulated genes (ISGs). The higher levels of viral replication and antitumor activity of oncoVV-WCL were further demonstrated in a mouse xenograft tumor model. Therefore, the engineered oncoVV expressing WCL might provide a new avenue for anticancer gene therapy.


Asunto(s)
Antineoplásicos/farmacología , Lectinas/genética , Lectinas/farmacología , Virus Oncolíticos/genética , Trucha/genética , Virus Vaccinia/genética , Animales , Antineoplásicos/uso terapéutico , Antivirales/farmacología , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Femenino , Humanos , Factor 3 Regulador del Interferón/genética , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Ratones Endogámicos BALB C , Ratones Desnudos , Viroterapia Oncolítica , Virus Oncolíticos/crecimiento & desarrollo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Virus Vaccinia/crecimiento & desarrollo , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Lett ; 509: 26-38, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33819529

RESUMEN

Oncolytic adenovirus-mediated gene therapy shows promise for cancer treatment; however, the systemic delivery of oncolytic adenovirus to tumors remains challenging. Recently, mesenchymal stem cells (MSCs) have emerged as potential vehicles for improving delivery. Yet, because the oncolytic adenovirus replicates in MSCs, balancing MSC viability with viral load is key to achieving optimal therapeutic effect. We thus developed an all-in-one Tet-on system that can regulate replication of oncolytic adenovirus. Then, we loaded the novel oncolytic adenovirus carrying interleukin (IL)-24 and/or Endostatin in human umbilical cord blood-mesenchymal stem cells (hUCB-MSCs) for glioma therapy. In vitro assays demonstrated that this novel oncolytic adenovirus could efficiently replicate and kill glioma cells while sparing normal cells. Moreover, doxycycline effectively regulated oncolytic adenovirus replication in the hUCB-MSCs. The doxycycline induction group with dual expression of IL-24 and Endostatin exhibited significantly greater antitumor effects than other groups in a xenograft model of glioma. Thus, this strategy for systemic delivery of oncolytic adenovirus with its oncolytic activity controlled by a Tet-on system is a promising method for achieving antitumor efficacy in glioma, especially for metastatic tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Trasplante de Células Madre de Sangre del Cordón Umbilical , Endostatinas/biosíntesis , Terapia Genética , Glioma/terapia , Interleucinas/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/virología , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virología , Muerte Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Endostatinas/genética , Femenino , Vectores Genéticos , Glioma/genética , Glioma/metabolismo , Glioma/virología , Humanos , Interleucinas/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/crecimiento & desarrollo , Carga Tumoral , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Virus Res ; 292: 198232, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33207264

RESUMEN

Senecavirus A (SVA), previously known as Seneca Valley virus, is classified into the genus Senecavirus in the family Picornaviridae. SVA is not pathogenic to normal human cells, but has potent oncolytic activity in some tumor cells with neuroendocrine feature, such as small cell lung cancer (SCLC) NCI-H446 cell line. In this study, we rescued and characterized a recombinant SVA that could efficiently express a novel luciferase, NanoLuc® luciferase (NLuc), which was smaller and "brighter" than others. This NLuc-tagged recombinant SVA (rSVA-NLuc) exhibited high capacity for viral replication, but genetic instability of NLuc during serial virus passages. The NLuc as a reporter facilitated oncolytic analysis of rSVA-NLuc in H446 cells. The rSVA-NLuc-infected H446 cells exhibited an oncolytic phenotype characterized by cell rounding, swelling, detachment and lysis at 48 h post infection. Kinetic curve showed that the NLuc was rapidly expressed in H446 cells during an exponential phase of viral growth. Because the NLuc offered several advantages over fluorescent proteins for assay scalability in vivo, the rSVA-NLuc would play a potential role in facilitating in vivo imaging studies of oncolytic virotherapy.


Asunto(s)
Luciferasas/genética , Virus Oncolíticos/genética , Picornaviridae/genética , Línea Celular Tumoral , Expresión Génica , Humanos , Luciferasas/metabolismo , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/fisiología , Picornaviridae/crecimiento & desarrollo , Picornaviridae/fisiología , Replicación Viral
5.
Biotechnol J ; 15(4): e1900411, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31950598

RESUMEN

Recent clinical trials have shown the potential of oncolytic adenoviruses as a cancer immunotherapy. A successful transition of oncolytic adenovirus to clinical applications requires efficient and good manufacturing practice compatible production and purification bioprocesses. Suspension cultures are preferable for virus production as they can reduce process costs and increase product quality and consistency. This work describes the adaptation of the A549 cell line to suspension culture in serum-reduced medium validated by oncolytic adenovirus production in stirred tank bioreactor. Cell concentrations up to 3 × 106 cells mL-1 are obtained during the production process. At harvest 1.4 × 1010 infectious particles mL-1 and 6.9 ± 1.1 × 1010 viral genome mL-1 are obtained corresponding to a viral genome: infectious particles ratio of 5.2 (± 1.9): 1 confirming the virus quality. Overall, the suspension characteristics of these A549 cells support an easily scalable, less time-consuming, and more cost-effective process for expanded success in the use of oncolytic viruses for cancer therapy.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Técnicas de Cultivo de Célula/métodos , Virus Oncolíticos/crecimiento & desarrollo , Células A549 , Adenoviridae/genética , Reactores Biológicos , Medios de Cultivo , Genoma Viral , Humanos , Microscopía Electrónica de Transmisión , Virus Oncolíticos/genética , Suspensiones , Cultivo de Virus
6.
Methods Mol Biol ; 2097: 253-263, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31776931

RESUMEN

Developments in genetic engineering have allowed researchers and clinicians to begin harnessing viruses to target and kill cancer cells, either through direct lysis or through recruitment of antiviral immune responses. Two powerful viruses in the fight against cancer are the single-stranded RNA viruses vesicular stomatitis virus and Zika virus. Here, we describe methods to propagate and titer these two viruses. We also describe a simple cell-killing assay to begin testing modified viruses for increased potential killing of glioblastoma cells.


Asunto(s)
Viroterapia Oncolítica/métodos , Virus Oncolíticos/crecimiento & desarrollo , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo , Virus Zika/crecimiento & desarrollo , Animales , Línea Celular , Citotoxicidad Inmunológica , Humanos , Ensayo de Placa Viral
7.
J Virol ; 93(19)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31315994

RESUMEN

As many tumor cells synthetize vascular endothelial growth factors (VEGF) that promote neo-vascularization and metastasis, frontline cancer therapies often administer anti-VEGF (α-VEGF) antibodies. To target the oncolytic parvovirus minute virus of mice (MVM) to the tumor vasculature, we studied the functional tolerance, evasion of neutralization, and induction of α-VEGF antibodies of chimeric viruses in which the footprint of a neutralizing monoclonal antibody within the 3-fold capsid spike was replaced by VEGF-blocking peptides: P6L (PQPRPL) and A7R (ATWLPPR). Both peptides allowed viral genome replication and nuclear translocation of chimeric capsid subunits. MVM-P6L efficiently propagated in culture, exposing the heterologous peptide on the capsid surface, and evaded neutralization by the anti-spike monoclonal antibody. In contrast, MVM-A7R yielded low infectious titers and was poorly recognized by an α-A7R monoclonal antibody. MVM-A7R showed a deficient assembly pattern, suggesting that A7R impaired a transitional configuration that the subunits must undergo in the 3-fold axis to close up the capsid shell. The MVM-A7R chimeric virus consistently evolved in culture into a mutant carrying the P6Q amino acid substitution within the A7R sequence, which restored normal capsid assembly and infectivity. Consistent with this finding, anti-native VEGF antibodies were induced in mice by a single injection of MVM-A7R empty capsids, but not by MVM-A7R virions. This fundamental study provides insights to endow an infectious parvovirus with immune antineovascularization and evasion capacities by replacing an antibody footprint in the capsid 3-fold axis with VEGF-blocking peptides, and it also illustrates the evolutionary capacity of single-stranded DNA (ssDNA) viruses to overcome engineered capsid structural restrictions.IMPORTANCE Targeting the VEGF signaling required for neovascularization by vaccination with chimeric capsids of oncolytic viruses may boost therapy for solid tumors. VEGF-blocking peptides (VEbp) engineered in the capsid 3-fold axis endowed the infectious parvovirus MVM with the ability to induce α-VEGF antibodies without adjuvant and to evade neutralization by MVM-specific antibodies. However, these properties may be compromised by structural restraints that the capsid imposes on the peptide configuration and by misassembly caused by the heterologous peptides. Significantly, chimeric MVM-VEbp resolved the structural restrictions by selecting mutations within the engineered peptides that restored efficient capsid assembly. These data show the promise of antineovascularization vaccines using chimeric VEbp-icosahedral capsids of oncolytic viruses but also raise safety concerns regarding the genetic stability of manipulated infectious parvoviruses in cancer and gene therapies.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Virus Diminuto del Ratón/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Proteínas de la Cápside/genética , Ratones Endogámicos BALB C , Virus Diminuto del Ratón/genética , Virus Diminuto del Ratón/crecimiento & desarrollo , Virus Oncolíticos/genética , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Carga Viral , Ensamble de Virus , Acoplamiento Viral , Internalización del Virus
8.
Viruses ; 11(6)2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31216641

RESUMEN

The rat protoparvovirus H-1PV is nonpathogenic in humans, replicates preferentially in cancer cells, and has natural oncolytic and oncosuppressive activities. The virus is able to kill cancer cells by activating several cell death pathways. H-1PV-mediated cancer cell death is often immunogenic and triggers anticancer immune responses. The safety and tolerability of H-1PV treatment has been demonstrated in early clinical studies in glioma and pancreatic carcinoma patients. Virus treatment was associated with surrogate signs of efficacy including immune conversion of tumor microenvironment, effective virus distribution into the tumor bed even after systemic administration, and improved patient overall survival compared with historical control. However, monotherapeutic use of the virus was unable to eradicate tumors. Thus, further studies are needed to improve H-1PV's anticancer profile. In this review, we describe H-1PV's anticancer properties and discuss recent efforts to improve the efficacy of H-1PV and, thereby, the clinical outcome of H-1PV-based therapies.


Asunto(s)
Parvovirus H-1/crecimiento & desarrollo , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Viroterapia Oncolítica/tendencias , Virus Oncolíticos/crecimiento & desarrollo , Animales , Terapia Combinada/métodos , Humanos , Ratas , Resultado del Tratamiento
9.
J Virol ; 93(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31092575

RESUMEN

Oncolytic virotherapy represents a promising experimental anticancer strategy, based on the use of genetically modified viruses to selectively infect and kill cancer cells. Vesicular stomatitis virus (VSV) is a prototypic oncolytic virus (OV) that induces cancer cell death through activation of the apoptotic pathway, although intrinsic resistance to oncolysis is found in some cell lines and many primary tumors, as a consequence of residual innate immunity to the virus. In the effort to improve OV therapeutic efficacy, we previously demonstrated that different agents, including histone deacetylase inhibitors (HDIs), functioned as reversible chemical switches to dampen the innate antiviral response and improve the susceptibility of resistant cancer cells to VSV infection. In the present study, we demonstrated that the NAD+-dependent histone deacetylase SIRT1 (silent mating type information regulation 2 homolog 1) plays a key role in the permissivity of prostate cancer PC-3 cells to VSVΔM51 replication and oncolysis. HDI-mediated enhancement of VSVΔM51 infection and cancer cell killing directly correlated with a decrease of SIRT1 expression. Furthermore, pharmacological inhibition as well as silencing of SIRT1 by small interfering RNA (siRNA) was sufficient to sensitize PC-3 cells to VSVΔM51 infection, resulting in augmentation of virus replication and spread. Mechanistically, HDIs such as suberoylanilide hydroxamic acid (SAHA; Vorinostat) and resminostat upregulated the microRNA miR-34a that regulated the level of SIRT1. Taken together, our findings identify SIRT1 as a viral restriction factor that limits VSVΔM51 infection and oncolysis in prostate cancer cells.IMPORTANCE The use of nonpathogenic viruses to target and kill cancer cells is a promising strategy in cancer therapy. However, many types of human cancer are resistant to the oncolytic (cancer-killing) effects of virotherapy. In this study, we identify a host cellular protein, SIRT1, that contributes to the sensitivity of prostate cancer cells to infection by a prototypical oncolytic virus. Knockout of SIRT1 activity increases the sensitivity of prostate cancer cells to virus-mediated killing. At the molecular level, SIRT1 is controlled by a small microRNA termed miR-34a. Altogether, SIRT1 and/or miR-34a levels may serve as predictors of response to oncolytic-virus therapy.


Asunto(s)
Interacciones Microbiota-Huesped , Inmunidad Innata , Virus Oncolíticos/crecimiento & desarrollo , Sirtuina 1/metabolismo , Vesiculovirus/crecimiento & desarrollo , Replicación Viral , Humanos , Masculino , Virus Oncolíticos/inmunología , Células PC-3 , Vesiculovirus/inmunología
10.
Int J Mol Sci ; 20(3)2019 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-30709038

RESUMEN

Oncolytic adenoviruses can trigger lysis of tumor cells, induce an antitumor immune response, bypass classical chemotherapeutic resistance strategies of tumors, and provide opportunities for combination strategies. A major challenge is the development of scalable production methods for viral seed stocks and sufficient quantities of clinical grade viruses. Because of promising clinical signals in a compassionate use program (Advanced Therapy Access Program) which supported further development, we chose the oncolytic adenovirus ONCOS-401 as a testbed for a new approach to scale up. We found that the best viral production conditions in both T-175 flasks and HYPERFlasks included A549 cells grown to 220,000 cells/cm² (80% confluency), with ONCOS-401 infection at 30 multiplicity of infection (MOI), and an incubation period of 66 h. The Lysis A harvesting method with benzonase provided the highest viral yield from both T-175 and HYPERFlasks (10,887 ± 100 and 14,559 ± 802 infectious viral particles/cell, respectively). T-175 flasks and HYPERFlasks produced up to 2.1 × 108 ± 0.2 and 1.75 × 108 ± 0.08 infectious particles of ONCOS-401 per cm² of surface area, respectively. Our findings suggest a suitable stepwise process that can be applied to optimizing the initial production of other oncolytic viruses.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Virus Oncolíticos/crecimiento & desarrollo , Cultivo de Virus/instrumentación , Células A549 , Animales , Técnicas de Cultivo Celular por Lotes/instrumentación , Humanos , Carga Viral , Cultivo de Virus/métodos , Replicación Viral
11.
J Cell Physiol ; 234(6): 8636-8646, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30515798

RESUMEN

Cancer therapy using oncolytic viruses is an emerging area, in which viruses are engineered to selectively propagate in tumor tissues without affecting healthy cells. Because of the advantages that adenoviruses (Ads) have over other viruses, they are more considered. To achieve tumor selectivity, two main modifications on Ads genome have been applied: small deletions and insertion of tissue- or tumor-specific promoters. Despite oncolytic adenoviruses ability in tumor cell lysis and immune responses stimulation, to further increase their antitumor effects, genomic modifications have been carried out including insertion of checkpoint inhibitors and antigenic or immunostimulatory molecules into the adenovirus genome and combination with dendritic cells and chemotherapeutic agents. This study reviews oncolytic adenoviruses structures, their antitumor efficacy in combination with other therapeutic strategies, and finally challenges around this treatment approach.


Asunto(s)
Adenoviridae/patogenicidad , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Adenoviridae/genética , Adenoviridae/crecimiento & desarrollo , Adenoviridae/inmunología , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Quimioterapia Adyuvante , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Terapia Genética , Humanos , Inmunoterapia Adoptiva , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/virología , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Replicación Viral
12.
Virology ; 522: 244-259, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30055515

RESUMEN

Vesicular stomatitis virus (VSV) shows potential for targeting and killing cancer cells, but can be dangerous in the brain due to its neurotropic glycoprotein. Here we test a chimeric virus in which the VSV glycoprotein is replaced with the Chikungunya polyprotein E3-E2-6K-E1 (VSVΔG-CHIKV). Control mice with brain tumors survived a mean of 40 days after tumor implant. VSVΔG-CHIKV selectively infected and eliminated the tumor, and extended survival substantially in all tumor-bearing mice to over 100 days. VSVΔG-CHIKV also targeted intracranial primary patient derived melanoma xenografts. Virus injected into one melanoma spread to other melanomas within the same brain with little detectable infection of normal cells. Intravenous VSVΔG-CHIKV infected tumor cells but not normal tissue. In immunocompetent mice, VSVΔG-CHIKV selectively infected mouse melanoma cells within the brain. These data suggest VSVΔG-CHIKV can target and destroy brain tumors in multiple animal models without the neurotropism associated with the wild type VSV glycoprotein.


Asunto(s)
Neoplasias Encefálicas/terapia , Virus Chikungunya/crecimiento & desarrollo , Virus Chikungunya/genética , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/genética , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/genética , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Melanoma/terapia , Ratones , Trasplante de Neoplasias , Viroterapia Oncolítica , Recombinación Genética , Análisis de Supervivencia , Resultado del Tratamiento
14.
J Med Virol ; 90(10): 1669-1673, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29797583

RESUMEN

Oncolytic adenoviral immunotherapy activates the innate immune system with subsequent induction of adaptive tumor-specific immune responses to fight cancer. Hence, oncolytic viruses do not only eradicate cancer cells by direct lysis, but also generate antitumor immune response, allowing for long-lasting cancer control and tumor reduction. Their therapeutic effect can be further enhanced by arming the oncolytic adenovirus with costimulatory transgenes and/or coadministration with other antitumor therapies. ONCOS-102 has already been found to be well tolerated and efficacious against some types of treatment-refractory tumors, including mesothelin-positive ovarian cancer (NCT01598129). It induced local and systemic CD8+ T-cell immunity and upregulated programmed death ligand 1. These results strongly advocate the use of ONCOS-102 in combination with other therapeutic strategies in advanced and refractory tumors, especially those expressing the mesothelin antigen. The in vivo work presented herein describes the ability of the oncolytic adenovirus ONCOS-102 to induce mesothelin-specific T-cells after the administration of the virus in bagg albino (BALB/c) mice with mesothelin-positive tumors. We also demonstrate the effectiveness of the interferon-γ the enzyme-linked immunospot (ELISPOT) assay to detect the induction of T-cells recognizing mesothelin, hexon, and E1A antigens in ONCOS-102-treated mesothelioma-bearing BALB/c mice. Thus, the ELISPOT assay could be useful to monitor the progress of therapy with ONCOS-102.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Anticuerpos Antineoplásicos/sangre , Mesotelioma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Ensayo de Immunospot Ligado a Enzimas , Humanos , Mesotelina , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Resultado del Tratamiento
15.
Cell Death Dis ; 9(3): 274, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449555

RESUMEN

Muscle-invasive bladder cancer (MIBC) is associated with low survival and high recurrence rates even in cases in which patients receive systemic treatments, such as surgery and chemotherapy. Here, we found that a naturally existing alphavirus, namely, M1, selectively kills bladder cancer cells but not normal cells, findings supported by our observations of changes in viral replication and MIBC and patient-derived MIBC cell apoptosis. Transcriptome analysis revealed that interferon-stimulated genes (ISGs) are expressed at low levels in sensitive bladder cancer cells and high levels in resistant cells. Knocking down ZC3HAV1 (ZAP), an antiviral factor in ISGs, restores M1 virus reactivity in resistant cells, and overexpressing ZAP partially reverses M1 virus-induced decreases in cell viability in sensitive cells. In orthotopic MIBC mice, tail vein injections of M1 significant inhibit tumor growth and prolong survival period, antitumor effects of M1 are stronger than those of the first-line chemotherapy agent cisplatin (CDDP). Treated tumors display enhanced cleaved-caspase-3 signals, which are representative of cell apoptosis, and decreased Ki-67 signals, which are representative of cell proliferation. Moreover, tissue microarray (TMA) analyses of clinical tumor specimens revealed that up to 45.6% of cases of MIBC presented with low ZAP expression, a finding that is prevalent in advanced MIBC. Our results indicate that the oncolytic virus M1 is a novel agent capable of functioning as a precise and effective therapy for MIBC.


Asunto(s)
Alphavirus/patogenicidad , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Neoplasias de la Vejiga Urinaria/terapia , Anciano , Alphavirus/crecimiento & desarrollo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Femenino , Interacciones Huésped-Patógeno , Humanos , Antígeno Ki-67/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Virus Oncolíticos/crecimiento & desarrollo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Tiempo , Carga Tumoral , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/virología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Sci Rep ; 8(1): 2233, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396500

RESUMEN

Cancer-specific promoter driven replication of oncolytic adenovirus (Ad) is cancer-specific, but shows low transcriptional activity. Thus, we generated several chimeric α-fetoprotein (AFP) promoter variants, containing reconstituted enhancer and silencer regions, to preferentially drive Ad replication in hepatocellular carcinoma (HCC). Modified AFP promoter, containing 2 enhancer A regions and a single enhancer B region (a2bm), showed strong and HCC-specific transcription. In AFP-positive HCCs, gene expression was 43- to 456-fold higher than those of control AFP promoter lacking enhancers. a2bm promoter was further modified by inserting multiple hypoxia-responsive elements (HRE) to generate Ha2bm promoter, which showed stronger transcriptional activity than a2bm promoter under hypoxic conditions. Ha2bm promoter-regulated oncolytic Ad (Ha2bm-d19) showed a stronger antitumor and proapoptotic effect than did a2bm promoter-regulated oncolytic Ad (a2bm-d19) in HCC xenograft tumors. Systemically administered Ha2bm-d19 caused no observable hepatotoxicity, whereas control replication-competent Ad, lacking cancer specificity (d19), induced significant hepatic damage. Ha2bm-d19 caused significantly lower expression of interleukin-6 than d19, showing that HCC-targeted delivery of Ad attenuates induction of the innate immune response against Ad. This chimeric AFP promoter enabled Ad to overcome the hypoxic tumor microenvironment and target HCC with high specificity, rendering it a promising candidate for the treatment of aggressive HCCs.


Asunto(s)
Adenoviridae/genética , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Células A549 , Adenoviridae/crecimiento & desarrollo , Animales , Apoptosis/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral , Células HEK293 , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Desnudos , Virus Oncolíticos/crecimiento & desarrollo , Regiones Promotoras Genéticas/genética , Transcripción Genética/genética , Microambiente Tumoral
17.
J Virol Methods ; 251: 69-74, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982603

RESUMEN

Newcastle Disease Virus (NDV) is an avian paramyxovirus that has no significant pathogenicity in humans. Cancer cells with impaired immune defense mechanisms are susceptible to infection and lysis by NDV. A recombinant construct of a lentogenic form of NDV (rNDV) containing an insertion of granulocyte macrophage colony stimulating factor (GMCSF) transgene was earlier reported and shown to have acceptably low avian pathogenicity as well as oncolytic potential. Reliable measurement of infectious titer is key to determining the effectiveness of virus preparations to infect and lyse cells. We report here a comparative evaluation of two infectious titer assays as applied to rNDV: plaque assay and fluorescent focus assay (FFA). Optimization of assay conditions for both titer methods has produced concordant results spanning several orders of magnitude. While plaque formation is the gold standard measure of virus titer, FFA provides higher throughput and faster turn-around. FFA has been further evaluated on two different instrument platforms, for automated versus manual foci recognition and counting, with equivalent results. These results point to amenability of FFA to transfer between different laboratories and analysts, without introducing significant subjectivity in data analysis.


Asunto(s)
Virus de la Enfermedad de Newcastle/crecimiento & desarrollo , Virus Oncolíticos/crecimiento & desarrollo , Imagen Óptica/métodos , Carga Viral/métodos , Ensayo de Placa Viral/métodos , Animales , Humanos
18.
J Infect Dis ; 217(5): 721-730, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29228368

RESUMEN

Cells latently infected with human immunodeficiency virus (HIV) evade immune- and drug-mediated clearance. These cells harbor intracellular signaling defects, including impairment of the antiviral type I interferon response. Such defects have also been observed in several cancers and have been exploited for the development of therapeutic oncolytic viruses, including the recombinant Maraba virus (MG1). We therefore hypothesized that MG1 would infect and eliminate cells latently infected with HIV-1, while sparing healthy uninfected cells. Preferential infection and elimination by MG1 was first demonstrated in cell lines latently infected with HIV-1. Following this, a reduction in HIV-1 DNA and inducible HIV-1 replication was observed following MG1 infection of latently infected, resting CD4+ T cells generated using an in vitro model of latency. Last, MG1 infection resulted in a reduction in HIV-1 DNA and inducible HIV-1 replication in memory CD4+ T cells isolated from effectively treated, HIV-1-infected individuals. Our results therefore highlight a novel approach to eliminate the latent HIV-1 reservoir.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/virología , VIH-1/fisiología , Virus Oncolíticos/crecimiento & desarrollo , Vesiculovirus/crecimiento & desarrollo , Latencia del Virus , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Infecciones por VIH/terapia , Humanos
19.
Biotechnol Bioeng ; 115(5): 1186-1194, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29288575

RESUMEN

Oncolytic viruses offer new hope to millions of patients with incurable cancer. One promising class of oncolytic viruses is Measles virus, but its broad administration to cancer patients is currently hampered by the inability to produce the large amounts of virus needed for treatment (1010 -1012 virus particles per dose). Measles virus is unstable, leading to very low virus titers during production. The time of infection and time of harvest are therefore critical parameters in a Measles virus production process, and their optimization requires an accurate online monitoring system. We integrated a probe based on dielectric spectroscopy (DS) into a stirred tank reactor to characterize the Measles virus production process in adherent growing Vero cells. We found that DS could be used to monitor cell adhesion on the microcarrier and that the optimal virus harvest time correlated with the global maximum permittivity signal. In 16 independent bioreactor runs, the maximum Measles virus titer was achieved approximately 40 hr after the permittivity maximum. Compared to an uncontrolled Measles virus production process, the integration of DS increased the maximum virus concentration by more than three orders of magnitude. This was sufficient to achieve an active Measles virus concentration of > 1010 TCID50 ml-1 .


Asunto(s)
Espectroscopía Dieléctrica/métodos , Virus del Sarampión/crecimiento & desarrollo , Virus Oncolíticos/crecimiento & desarrollo , Tecnología Farmacéutica/métodos , Cultivo de Virus/métodos , Animales , Chlorocebus aethiops , Células Vero
20.
Viruses ; 9(12)2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29244745

RESUMEN

Glioblastoma, one of the most aggressive primary brain tumors, is characterized by highly immunosuppressive microenvironment. This contributes to glioblastoma resistance to standard treatment modalities and allows tumor growth and recurrence. Several immune-targeted approaches have been recently developed and are currently under preclinical and clinical investigation. Oncolytic viruses, including the autonomous protoparvovirus H-1 (H-1PV), show great promise as novel immunotherapeutic tools. In a first phase I/IIa clinical trial (ParvOryx01), H-1PV was safe and well tolerated when locally or systemically administered to recurrent glioblastoma patients. The virus was able to cross the blood-brain (tumor) barrier after intravenous infusion. Importantly, H-1PV treatment of glioblastoma patients was associated with immunogenic changes in the tumor microenvironment. Tumor infiltration with activated cytotoxic T cells, induction of cathepsin B and inducible nitric oxide (NO) synthase (iNOS) expression in tumor-associated microglia/macrophages (TAM), and accumulation of activated TAM in cluster of differentiation (CD) 40 ligand (CD40L)-positive glioblastoma regions was detected. These are the first-in-human observations of H-1PV capacity to switch the immunosuppressed tumor microenvironment towards immunogenicity. Based on this pilot study, we present a tentative model of H-1PV-mediated modulation of glioblastoma microenvironment and propose a combinatorial therapeutic approach taking advantage of H-1PV-induced microglia/macrophage activation for further (pre)clinical testing.


Asunto(s)
Glioblastoma/terapia , Parvovirus H-1/crecimiento & desarrollo , Factores Inmunológicos , Microglía/inmunología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/crecimiento & desarrollo , Linfocitos T Citotóxicos/inmunología , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Humanos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...