Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 13(6)2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071585

RESUMEN

Although the oncolytic parvovirus H-1PV has entered clinical trials, predicting therapeutic success remains challenging. We investigated whether the antiviral state in tumor cells determines the parvoviral oncolytic efficacy. The interferon/interferon-stimulated genes (IFN/ISG)-circuit and its major configurator, human endogenous retroviruses (HERVs), were evaluated using qRT-PCR, ELISA, Western blot, and RNA-Seq techniques. In pancreatic cancer cell lines, H-1PV caused a late global shutdown of innate immunity, whereby the concomitant inhibition of HERVs and IFN/ISGs was co-regulatory rather than causative. The growth-inhibitory IC50 doses correlated with the power of suppression but not with absolute ISG levels. Moreover, H-1PV was not sensitive to exogenous IFN despite upregulated antiviral ISGs. Such resistance questioned the biological necessity of the oncotropic ISG-shutdown, which instead might represent a surrogate marker for personalized oncolytic efficacy. The disabled antiviral homeostasis may modify the activity of other viruses, as demonstrated by the reemergence of endogenous AluY-retrotransposons. This way of suppression may compromise the interferogenicity of drugs having gemcitabine-like mechanisms of action. This shortcoming in immunogenic cell death induction is however amendable by immune cells which release IFN in response to H-1PV.


Asunto(s)
Parvovirus H-1/inmunología , Parvovirus H-1/patogenicidad , Homeostasis/inmunología , Inmunidad Innata , Interferones/inmunología , Neoplasias Pancreáticas/virología , Muerte Celular/inmunología , Línea Celular Tumoral , Citocinas , Humanos , Leucocitos Mononucleares/virología , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Virus Oncolíticos/patogenicidad , Infecciones por Parvoviridae/complicaciones , Infecciones por Parvoviridae/virología
2.
Methods Mol Biol ; 2058: 295-306, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31486047

RESUMEN

Fluorescence in situ hybridization (FISH) is a specific, sensitive, accurate, and reliable technique widely applied in both research and clinic. Here we describe the detailed protocol of a FISH method established by us to serve the scientific purposes of the first oncolytic parvovirus clinical trial (ParvOryx01). This trial was launched in Germany in 2011. After trial completion in 2015, results were published in Molecular Therapy in 2017. The primary purpose of the trial was to evaluate the safety of an oncolytic parvovirus, H-1PV (ParvOryx), in recurrent glioblastoma patients. In addition, the efficiency of H-1PV tumor targeting after intratumoral or systemic virus administration was assessed by FISH detection of viral nucleic acids (genomic single-stranded DNA, mRNA and parvovirus double-stranded replicative forms) in formalin-fixed paraffin-embedded glioblastoma tissues resected at day 10 after ParvOryx treatment. The FISH method allowed the detection-for the first time in humans-of H-1PV replication markers in brain tumors of parvovirus-treated patients. A protocol combining mRNA FISH with simultaneous immunofluorescent staining for tumor and tumor microenvironment markers was also developed and is described here, in order to better characterize H-1PV cellular targets and H-1PV treatment-associated tumor microenvironment changes.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , ADN Viral , Vectores Genéticos , Parvovirus H-1 , Hibridación Fluorescente in Situ , Virus Oncolíticos , Neoplasias Encefálicas/terapia , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Parvovirus H-1/genética , Parvovirus H-1/inmunología , Humanos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Microambiente Tumoral , Replicación Viral
3.
Front Immunol ; 10: 1848, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31440242

RESUMEN

Cancer cells utilize multiple mechanisms to evade and suppress anticancer immune responses creating a "cold" immunosuppressive tumor microenvironment. Oncolytic virotherapy is emerging as a promising approach to revert tumor immunosuppression and enhance the efficacy of other forms of immunotherapy. Growing evidence indicates that oncolytic viruses (OVs) act in a multimodal fashion, inducing immunogenic cell death and thereby eliciting robust anticancer immune responses. In this review, we summarize information about OV-mediated immune conversion of the tumor microenvironment. As a case study we focus on the rodent protoparvovirus H-1PV and its dual role as an oncolytic and immune modulatory agent. Potential strategies to improve H-1PV anticancer efficacy are also discussed.


Asunto(s)
Parvovirus H-1/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Microambiente Tumoral/inmunología , Humanos
4.
BMC Cancer ; 13: 367, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23902851

RESUMEN

BACKGROUND: Investigating how the immune system functions during malignancies is crucial to developing novel therapeutic strategies. Natural killer (NK) cells, an important component of the innate immune system, play a vital role in immune defense against tumors and virus-infected cells. The poor survival rate in colon cancer makes it particularly important to develop novel therapeutic strategies. Oncolytic viruses, in addition to lysing tumor cells, may have the potential to augment antitumor immune responses. In the present study, we investigate the role of NK cells and how parvovirus H-1PV can modulate NK-cell mediated immune responses against colon carcinoma. METHODS: Human NK cells were isolated from the blood of healthy donors. The cytotoxicity and antibody-mediated inhibition of NK cells were measured in chromium release assays. Phenotypic assessment of colon cancer and dendritic cells was done by FACS. The statistical significance of the results was calculated with Student's t test (*p <0.05; **, p < 0.01; ***, p < 0.001). RESULTS: We show that IL-2-activated human NK cells can effectively kill colon carcinoma cells. Killing of colon carcinoma cells by NK cells was further enhanced upon infection of the former cells with parvovirus H-1PV. H-1PV has potent oncolytic activity against various tumors, yet its direct killing effect on colon carcinoma cells is limited. The cytotoxicity of NK cells towards colon carcinoma cells, both mock- and H-1PV-infected, was found to be mostly mediated by a combination of natural cytotoxicity receptors (NCRs), namely NKp30, 44, and 46. Colon carcinoma cells displayed low to moderate expression of NK cell ligands, and this expression was modulated upon H-1PV infection. Lysates of H-1PV-infected colon carcinoma cells were found to increase MHC class II expression on dendritic cells. CONCLUSIONS: Altogether, these data suggest that IL-2-activated NK cells actively kill colon carcinoma cells and that this killing is mediated by several natural cytotoxicity receptors (NCRs) in combination. Additionally, in association with parvovirus H-1PV, IL-2-activated NK cells have the potential to boost immune responses against colon cancer.


Asunto(s)
Adenocarcinoma/inmunología , Neoplasias del Colon/inmunología , Citotoxicidad Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Adenocarcinoma/virología , Neoplasias del Colon/virología , Citometría de Flujo , Parvovirus H-1/inmunología , Humanos , Interleucina-2/inmunología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Infecciones por Parvoviridae/inmunología
5.
Int J Cancer ; 132(11): 2548-56, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23151948

RESUMEN

This study aimed to investigate the function of toll-like receptors (TLRs) during oncolytic parvovirus H-1 (H-1PV)-induced human immune responses. First, the role of TLRs in the activation of the NFκB transcription factor was characterized; second, the immunologic effects of H-1PV-induced tumor cell lysates (TCL) on human antitumor immune responses were evaluated. A human ex vivo model was used to study immune responses with dendritic cells (DCs). Human embryonic kidney cells (HEK293) transfected to stably express TLRs were used as potential human DC equivalents to further investigate the role of specific TLRs during immune activation. TLR3 and TLR9 were activated by H-1PV infection, which correlated with NFκB translocation to the nucleus and a reduced cytoplasmic IκB expression. Using a TLR-signaling reporter plasmid (pNiFty-Luc), NFκB activity was increased following H-1PV infection. In addition, human DCs coincubated with H-1PV-induced TCL demonstrated increased TLR3 and TLR9 expression. These data suggest that H-1PV-induced TCL stimulate human DCs at least in part through TLR-dependent signaling pathways. Thus, DC maturation occurred through exposure to H-1PV-induced TCL through TLR-signaling leading to NFκB-dependent activation of the adaptive immune system as indicated by the increased expression of CD86, TLR3 and TLR9. Furthermore, the transcription of various cytokines indicates the activation of immune response, therefore the production of the proinflammatory cytokine TNF-α was determined. Here, H-1PV-induced TCL significantly enhanced the TNF-α level by DCs after coculture. H-1PV oncolytic virotherapy enhances immune priming by different effects on DCs and generates antitumor immunity. These findings potentially offer a new approach to tumor therapy.


Asunto(s)
Células Dendríticas/inmunología , Parvovirus H-1/inmunología , Melanoma/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Infecciones por Parvoviridae/inmunología , Receptores Toll-Like/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citoplasma/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/patología , Células Dendríticas/virología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Sistema Inmunológico , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Melanoma/metabolismo , Melanoma/terapia , FN-kappa B/genética , FN-kappa B/metabolismo , Infecciones por Parvoviridae/patología , Infecciones por Parvoviridae/virología , Transducción de Señal , Receptores Toll-Like/inmunología , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología
6.
J Virol ; 86(14): 7554-64, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22553326

RESUMEN

An in-frame, 114-nucleotide-long deletion that affects the NS-coding sequence was created in the infectious molecular clone of the standard parvovirus H-1PV, thereby generating Del H-1PV. The plasmid was transfected and further propagated in permissive human cell lines in order to analyze the effects of the deletion on virus fitness. Our results show key benefits of this deletion, as Del H-1PV proved to exhibit (i) higher infectivity (lower particle-to-infectivity ratio) in vitro and (ii) enhanced tumor growth suppression in vivo compared to wild-type H-1PV. This increased infectivity correlated with an accelerated egress of Del H-1PV progeny virions in producer cells and with an overall stimulation of the viral life cycle in subsequently infected cells. Indeed, virus adsorption and internalization were significantly improved with Del H-1PV, which may account for the earlier appearance of viral DNA replicative forms that was observed with Del H-1PV than wild-type H-1PV. We hypothesize that the internal deletion within the NS2 and/or NS1 protein expressed by Del H-1PV results in the stimulation of some step(s) of the viral life cycle, in particular, a maturation step(s), leading to more efficient nuclear export of infectious viral particles and increased fitness of the virus produced.


Asunto(s)
Parvovirus H-1/genética , Parvovirus H-1/patogenicidad , Infecciones por Parvoviridae/patología , Eliminación de Secuencia , Proteínas no Estructurales Virales/genética , Línea Celular , Parvovirus H-1/inmunología , Humanos , Infecciones por Parvoviridae/virología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Acoplamiento Viral , Internalización del Virus , Liberación del Virus
7.
PLoS One ; 7(2): e32197, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359669

RESUMEN

BACKGROUND: H-1 parvovirus (H-1 PV), a rodent autonomous oncolytic parvovirus, has emerged as a novel class of promising anticancer agents, because of its ability to selectively find and destroy malignant cells. However, to probe H-1 PV multimodal antitumor potential one of the major prerequisites is to decipher H-1 PV direct interplay with human immune system, and so prevent any risk of impairment. METHODOLOGY/PRINCIPAL FINDINGS: Non activated peripheral blood mononuclear cells (PBMCs) are not sensitive to H-1 PV cytotoxic effect. However, the virus impairs both activated PBMC proliferation ability and viability. This effect is related to H-1 PV infection as evidenced by Western blotting detection of H-1 PV main protein NS1. However, TCID50 experiments did not allow newly generated virions to be detected. Moreover, flow cytometry has shown that H-1 PV preferentially targets B lymphocytes. Despite seeming harmful at first sight, H-1 PV seems to affect very few NK cells and CD8+ T lymphocytes and, above all, clearly does not affect human neutrophils and one of the major CD4+ T lymphocyte subpopulation. Very interestingly, flow cytometry analysis and ELISA assays proved that it even activates human CD4+ T cells by increasing activation marker expression (CD69 and CD30) and both effective Th1 and Th2 cytokine secretion (IL-2, IFN-γ and IL-4). In addition, H-1 PV action does not come with any sign of immunosuppressive side effect. Finally, we have shown the efficiency of H-1 PV on xenotransplanted human nasopharyngeal carcinoma, in a SCID mouse model reconstituted with human PBMC. CONCLUSIONS/SIGNIFICANCE: Our results show for the first time that a wild-type oncolytic virus impairs some immune cell subpopulations while directly activating a Helper CD4+ T cell response. Thus, our data open numerous gripping perspectives of investigation and strongly argue for the use of H-1 PV as an anticancer treatment.


Asunto(s)
Terapia Biológica/métodos , Linfocitos T CD4-Positivos/virología , Parvovirus H-1/inmunología , Virus Oncolíticos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Carcinoma , Humanos , Ratones , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/terapia , Linfocitos T Reguladores , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Biol Ther ; 10(12): 1280-9, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21124075

RESUMEN

Treatment of cancers by means of viruses, that specifically replicate in (oncotropism) and kill (oncolysis) neoplastic cells, is increasingly gaining acceptance in the clinic. Among these agents, parvoviruses have been shown to possess not only direct oncolytic but also immunomodulating properties, serving as an adjuvant to prime the immune system to react against infected tumors. Here, we aimed to establish whether immunomodulating mechanisms participate in the recently reported therapeutic potential of parvoviruses against pancreatic carcinoma. Using adoptive transfer experiments we discovered that the transfer of splenocytes of donor rats harboring H-1PV-treated orthotopic PDAC tumors could significantly prolong the survival of naïve tumor-bearing recipients, compared to those receiving cells from mock-treated donors. Closer investigation of immunological parameters in infected donor rats revealed that virus-induced interferon gamma production and cellular immune response played an important role in this effect. These data have also preclinical relevance since abortive H-1PV infection of human peripheral blood mononuclear cells or cocultivation of these cells with H-1PV-preinfected pancreatic cancer cells, resulted in enhancement of innate and adaptive immune reactivity. Taken together our data reveal that oncolytic H-1PV modulates the immune system into an anticancer state, and further support the concept of using parvoviruses in the fight against pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/terapia , Parvovirus H-1/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Viroterapia Oncolítica/métodos , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Citometría de Flujo , Parvovirus H-1/fisiología , Humanos , Inmunidad Celular/inmunología , Inmunomodulación , Interferón gamma/inmunología , Virus Oncolíticos , Reacción en Cadena de la Polimerasa , Ratas , Bazo/inmunología , Bazo/virología , Balance Th1 - Th2
9.
Oncol Rep ; 17(6): 1493-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487410

RESUMEN

Oncolytic viruses have emerged as a novel class of potent anticancer agents offering an improvement on chemo- and radiotherapy in terms of tumor targeting and reduction of side-effects. Among these agents, autonomous parvoviruses have attracted the attention of researchers for their ability to preferentially replicate in and kill transformed cells, and to suppress tumors in the absence of adverse reactions in various animal models. We have previously shown that lethally irradiated autologous tumor cells can support parvovirus H-1PV production and serve as carriers to deliver progeny H-1PV into the vicinity of lung metastases in a rat tumor model, resulting in H-1PV infection of and multiplication in metastatic cells. It is known that irradiated autologous (neoplastic) cells can also act as a therapeutic vaccine against the original tumor. Yet the ability of these cells to suppress metastases in the above model was found to be much increased as a result of their H-1PV infection. This prompted us to determine whether H-1PV boosted the tumor-suppressing capacity of the autologous vaccine by increasing its immunogenic potential and/or by making it a factory of oncolytic viruses able to reach and destroy the metastases. Both effects could be dissociated in the presence of neutralising antibodies which either prevent the progeny viruses from spreading to metastatic cells, or deplete the CD8 effector cells from the immune system. This strategy revealed that the H-1PV infection of tumor cells enhanced their ability to trigger an immune response for which uninfected tumor cells could be the targets, thereby amplifying and taking over from the direct viral oncolytic activity. This dual oncolytic/vaccinal effect of H-1PV holds out promises of clinical applications to cancer therapy.


Asunto(s)
Vacunas contra el Cáncer , Parvovirus H-1/fisiología , Neoplasias Pulmonares/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Animales , Anticuerpos Antivirales/farmacología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Parvovirus H-1/efectos de los fármacos , Parvovirus H-1/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Virus Oncolíticos/efectos de los fármacos , Ratas , Ratas Endogámicas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA