Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Viruses ; 16(4)2024 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-38675903

RESUMEN

Gliomas account for approximately 75-80% of all malignant primary tumors in the central nervous system (CNS), with glioblastoma multiforme (GBM) considered the deadliest. Despite aggressive treatment involving a combination of chemotherapy, radiotherapy, and surgical intervention, patients with GBM have limited survival rates of 2 to 5 years, accompanied by a significant decline in their quality of life. In recent years, novel management strategies have emerged, such as immunotherapy, which includes the development of vaccines or T cells with chimeric antigen receptors, and oncolytic virotherapy (OVT), wherein wild type (WT) or genetically modified viruses are utilized to selectively lyse tumor cells. In vitro and in vivo studies have shown that the Zika virus (ZIKV) can infect glioma cells and induce a robust oncolytic activity. Consequently, interest in exploring this virus as a potential oncolytic virus (OV) for high-grade gliomas has surged. Given that ZIKV actively circulates in Colombia, evaluating its neurotropic and oncolytic capabilities holds considerable national and international importance, as it may emerge as an alternative for treating highly complex gliomas. Therefore, this literature review outlines the generalities of GBM, the factors determining ZIKV's specific tropism for nervous tissue, and its oncolytic capacity. Additionally, we briefly present the progress in preclinical studies supporting the use of ZIKV as an OVT for gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Viroterapia Oncolítica , Virus Oncolíticos , Virus Zika , Animales , Humanos , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Glioblastoma/terapia , Glioblastoma/virología , Glioma/terapia , Glioma/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Virus Zika/fisiología , Infección por el Virus Zika/virología
2.
Viruses ; 16(3)2024 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-38543739

RESUMEN

The absence of tumor-infiltrating lymphocytes negatively impacts the response to chemotherapy and prognosis in all subtypes of breast cancer. Therapies that stimulate a proinflammatory environment may help improve the response to standard treatments and also to immunotherapies such as checkpoint inhibitors. Newcastle disease virus (NDV) shows oncolytic activity, as well as immune modulating potential, in the treatment of breast cancer in vitro and in vivo; however, its potential to enhance tumor-infiltrating immune cells in breast cancer has yet to be evaluated. Since spontaneous canine mammary tumors represent a translational model of human breast cancer, we conducted this proof-of-concept study, which could provide a rationale for further investigating NDV-MLS as immunotherapy for mammary cancer. Six female companion dogs with spontaneous mammary cancer received a single intravenous and intratumoral injection of oncolytic NDV-MLS. Immune cell infiltrates were evaluated by histology and immunohistochemistry in the stromal, intratumoral, and peritumoral compartments on day 6 after viral administration. Increasing numbers of immune cells were documented post-viral treatment, mainly in the peritumoral compartment, where plasma cells and CD3+ and CD3-/CD79- lymphocytes predominated. Viral administration was well tolerated, with no significant adverse events. These findings support additional research on the use of NDV-MLS immunotherapy for mammary cancer.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Animales , Femenino , Perros , Virus de la Enfermedad de Newcastle/fisiología , Mascotas , Virus Oncolíticos/fisiología , Inmunoterapia , Línea Celular Tumoral , Neoplasias/terapia
3.
Mol Ther ; 32(2): 440-456, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38213031

RESUMEN

Here we introduce a first-in-class microRNA-sensitive oncolytic Zika virus (ZIKV) for virotherapy application against central nervous system (CNS) tumors. The described methodology produced two synthetic modified ZIKV strains that are safe in normal cells, including neural stem cells, while preserving brain tropism and oncolytic effects in tumor cells. The microRNA-sensitive ZIKV introduces genetic modifications in two different virus sites: first, in the established 3'UTR region, and secondly, in the ZIKV protein coding sequence, demonstrating for the first time that the miRNA inhibition systems can be functional outside the UTR RNA sites. The total tumor remission in mice bearing human CNS tumors, including metastatic tumor growth, after intraventricular and systemic modified ZIKV administration, confirms the promise of this virotherapy as a novel agent against brain tumors-highly deadly diseases in urgent need of effective advanced therapies.


Asunto(s)
Neoplasias del Sistema Nervioso Central , MicroARNs , Viroterapia Oncolítica , Virus Oncolíticos , Infección por el Virus Zika , Virus Zika , Humanos , Ratones , Animales , Virus Oncolíticos/genética , Virus Zika/genética , MicroARNs/genética , Infección por el Virus Zika/terapia , Viroterapia Oncolítica/métodos
4.
Int J Mol Sci ; 24(12)2023 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-37373140

RESUMEN

More than one million women are diagnosed annually worldwide with a gynecological cancer. Most gynecological cancers are diagnosed at a late stage, either because a lack of symptoms, such as in ovarian cancer or limited accessibility to primary prevention in low-resource countries, such as in cervical cancer. Here, we extend the studies of AR2011, a stroma-targeted and tumor microenvironment responsive oncolytic adenovirus (OAdV), whose replication is driven by a triple hybrid promoter. We show that AR2011 was able to replicate and lyse in vitro fresh explants obtained from human ovarian cancer, uterine cancer, and cervical cancer. AR2011 was also able to strongly inhibit the in vitro growth of ovarian malignant cells obtained from human ascites fluid. The virus could synergize in vitro with cisplatin even on ascites-derived cells obtained from patients heavily pretreated with neoadjuvant chemotherapy. AR2011(h404), a dual transcriptionally targeted derived virus armed with hCD40L and h41BBL under the regulation of the hTERT promoter, showed a strong efficacy in vivo both on subcutaneous and intraperitoneally established human ovarian cancer in nude mice. Preliminary studies in an immunocompetent murine tumor model showed that AR2011(m404) expressing the murine cytokines was able to induce an abscopal effect. The present studies suggest that AR2011(h404) is a likely candidate as a novel medicine for intraperitoneal disseminated ovarian cancer.


Asunto(s)
Infecciones por Adenoviridae , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Ováricas , Neoplasias del Cuello Uterino , Femenino , Humanos , Ratones , Animales , Adenoviridae/genética , Ascitis , Ratones Desnudos , Microambiente Tumoral , Línea Celular Tumoral , Neoplasias Ováricas/terapia , Neoplasias Ováricas/tratamiento farmacológico , Virus Oncolíticos/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
PLoS One ; 18(5): e0285543, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37186587

RESUMEN

Despite advances in biomedical research, gastric cancer remains the leading cause of morbidity and mortality worldwide due to the limited efficacy of conventional therapies. In recent decades, oncolytic viruses have emerged as a biological therapeutic alternative to cancer due to their selectivity, effectiveness, and low toxicity. However, clinical trials have shown that developing a virus with selectivity for multiple tumor receptors and the ability to penetrate and diffuse through the tumor microenvironment to reactivate the immune system remains challenging. This study aimed to examine the oncolytic potential of tumor cell-adapted rotavirus Wt1-5 in gastric adenocarcinoma samples. This study focused on determining the propagation capacity of the RV Wt1-5 through the tumor and the importance of the expression of cell surface co-receptors, including integrin ß3, protein disulfide isomerase (PDI), and heat shock proteins (Hsp-90, -70, -60, -40, and Hsc 70), during infection of tumor cells. These proteins were found to be differentially expressed in tumor cells compared to adjacent non-tumor cells. Preincubation of gastric tumor cells with antibodies against these proteins decreased rotavirus infections, validating their importance in the binding and entry of RV Wt1-5 into tumor cells, as previously reported. Upon RV infection, apoptosis was one of the types of death that was observed. This was evidenced by evaluating the expression of CASP-3, -9, PARP, cytochrome C, Bax, Bid, p53, and Bcl-2, as well as observing morphological changes such as chromatin margination, nuclear condensation, and fragmentation. Finally, at 60 h.p.i, histological analysis revealed that oncolysis compromised the entire thickness of the tumor. Therefore, the results suggest that RV Wt1-5 could be a novel therapeutic agent co-adjuvant agent for conventional and targeted therapies in managing GC. Ex vivo infection of the tumor tissue model showed characteristics of an immune response that could be explored in future studies.


Asunto(s)
Adenocarcinoma , Viroterapia Oncolítica , Virus Oncolíticos , Infecciones por Rotavirus , Rotavirus , Neoplasias Gástricas , Humanos , Rotavirus/fisiología , Neoplasias Gástricas/terapia , Virus Oncolíticos/fisiología , Adenocarcinoma/terapia , Viroterapia Oncolítica/métodos , Microambiente Tumoral
6.
DNA Cell Biol ; 42(6): 265-266, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37163221
7.
Clin Transl Oncol ; 24(9): 1682-1701, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35612653

RESUMEN

With the in-depth research and wide application of immunotherapy recently, new therapies based on oncolytic viruses are expected to create new prospects for cancer treatment via eliminating the suppression of the immune system by tumors. Currently, an increasing number of viruses are developed and engineered, and various virus vectors based on effectively stimulating human immune system to kill tumor cells have been approved for clinical treatment. Although the virus can retard the proliferation of tumor cells, the choice of oncolytic viruses in biological cancer therapy is equally critical given their therapeutic efficacy, safety and adverse effects. Moreover, previously known oncolytic viruses have not been systematically classified. Therefore, in this review, we summarized and distinguished the characteristics of several common types of oncolytic viruses: herpes simplex virus, adenovirus, measles virus, Newcastle disease virus, reovirus and respiratory syncytial virus. Subsequently, we outlined that these oncolytic viral vectors have been transformed from preclinical studies in combination with immunotherapy, radiotherapy, chemotherapy, and nanoparticles into clinical therapeutic strategies for various advanced solid malignancies or circulatory system cancers.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Vectores Genéticos , Humanos , Inmunoterapia , Neoplasias/terapia , Virus Oncolíticos/genética
8.
J Thorac Cardiovasc Surg ; 163(4): e313-e328, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33485667

RESUMEN

OBJECTIVE: The mainstay of treatment for patients with malignant pleural disease is fluid drainage and systemic therapy. A tumor-specific oncolytic virus or T-cell-activating interleukin-2 immunotherapy may provide an opportunity for local control. We previously developed a vaccinia virus-expressing interleukin-2, an oncolytic virus that mediated tumor regression in preclinical peritoneal tumor models with expansion of tumor-infiltrating lymphocytes. We evaluated the antitumor efficacy and immune modulatory effects of vaccinia virus-expressing interleukin-2 in malignant pleural disease. METHODS: A murine model of malignant pleural disease was established with percutaneous intrapleural deposition of the Lewis lung carcinoma cell line and monitored with bioluminescent imaging. After intrapleural or systemic administration of vaccinia viruses (vaccinia virus yellow fluorescent protein control, vaccinia virus-expressing interleukin-2), systemic anti-programmed cell death-1 antibody, or combination therapy (vaccinia virus-expressing interleukin-2 and anti-programmed cell death-1), tumor mass, immune cell infiltration, T-cell receptor diversity, and survival were assessed. RESULTS: Intrapleural vaccinia virus resulted in significant tumor regression compared with phosphate-buffered saline control (P < .05). Inclusion of the interleukin-2 transgene further increased intratumoral CD8+ T cells (P < .01) and programmed cell death-1 expression on CD8+ tumor-infiltrating lymphocytes (P < .001). Intrapleural vaccinia virus-expressing interleukin-2 was superior to systemic vaccinia virus-expressing interleukin-2, with reduced tumor burden (P < .0001) and improved survival (P < .05). Intrapleural vaccinia virus-expressing interleukin-2 alone or combined treatment with systemic anti-programmed cell death-1 reduced tumor burden (P < .01), improved survival (P < .01), and increased intratumoral αß T-cell receptor diversity (P < .05) compared with systemic anti-programmed cell death-1 monotherapy. CONCLUSIONS: Intrapleural vaccinia virus-expressing interleukin-2 reduced tumor burden and enhanced survival in a murine malignant pleural disease model. Increased CD8+ tumor-infiltrating lymphocytes and αß T-cell receptor diversity are associated with enhanced response. Clinical trials will enable assessment of intrapleural vaccinia virus-expressing interleukin-2 therapy in patients with malignant pleural disease.


Asunto(s)
Interleucina-2/metabolismo , Neoplasias Pulmonares/inmunología , Viroterapia Oncolítica , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/metabolismo , Virus Vaccinia
9.
Viruses ; 13(10)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34696533

RESUMEN

The Zika virus (ZIKV) has shown a promising oncolytic effect against embryonal CNS tumors. However, studies on the effect of different administration routes and the ideal viral load in preclinical models are highly relevant aiming for treatment safety and efficiency. Here, we investigated the effect and effectiveness of different routes of administration, and the number of ZIKVBR injections on tumor tropism, destruction, and side effects. Furthermore, we designed an early-stage human brain organoid co-cultured with embryonal CNS tumors to analyze the ZIKVBR oncolytic effect. We showed that in the mice bearing subcutaneous tumors, the ZIKVBR systemically presented a tropism to the brain. When the tumor was located in the mice's brain, serial systemic injections presented efficient tumor destruction, with no neurological or other organ injury and increased mice survival. In the human cerebral organoid model co-cultured with embryonal CNS tumor cells, ZIKVBR impaired tumor progression. The gene expression of cytokines and chemokines in both models suggested an enhancement of immune cells recruitment and tumor inflammation after the treatment. These results open new perspectives for virotherapy using the ZIKVBR systemic administration route and multiple doses of low virus load for safe and effective treatment of embryonal CNS tumors, an orphan disease that urges new effective therapies.


Asunto(s)
Neoplasias Encefálicas/terapia , Viroterapia Oncolítica/métodos , Virus Zika/metabolismo , Animales , Encéfalo/virología , Neoplasias Encefálicas/patología , Línea Celular , Sistema Nervioso Central/efectos de los fármacos , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Inmunoterapia/métodos , Inyecciones Intralesiones/métodos , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Virus Oncolíticos/metabolismo , Organoides , Virus Zika/inmunología , Infección por el Virus Zika/virología
10.
J Immunol Res ; 2021: 3412906, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34557553

RESUMEN

Glioblastoma (GBM) is the most common primary malignant brain tumor with a high mortality rate. The current treatment consists of surgical resection, radiation, and chemotherapy; however, the median survival rate is only 12-18 months despite these alternatives, highlighting the urgent need to find new strategies. The heterogeneity of GBM makes this tumor difficult to treat, and the immunotherapies result in an attractive approach to modulate the antitumoral immune responses favoring the tumor eradication. The immunotherapies for GMB including monoclonal antibodies, checkpoint inhibitors, vaccines, and oncolytic viruses, among others, have shown favorable results alone or as a multimodal treatment. In this review, we summarize and discuss promising immunotherapies for GBM currently under preclinical investigation as well as in clinical trials.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunoterapia , Animales , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores de Tumor , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Terapia Combinada , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Glioblastoma/etiología , Glioblastoma/metabolismo , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Modelos Animales , Terapia Molecular Dirigida , Viroterapia Oncolítica/métodos , Resultado del Tratamiento
11.
Vet Comp Oncol ; 19(3): 593-601, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33871162

RESUMEN

Canine mammary carcinoma (CMC) is one of the major health threats in dogs. The oncolytic virotherapy is a promising strategy to treat canine as well as human cancer patients with non-pathogenic replicating viruses. Here, we evaluated the antitumor activity of one lentogenic, non-lytic Newcastle disease virus (NDV) LaSota strain expressing GFP (NDV-GFP) on five different CMCs and one non-tumorigenic cell line, regarding cell viability, cell death, selectivity index, morphology, global and target gene expression analysis. As evidenced by the selectivity index, all CMC cell lines were more susceptible to NDV-GFP in comparison with the non-tumorigenic cells (~3.1× to ~78.7×). In addition, the oncolytic effect of NDV-GFP was more evident in more malignant CMC cells. Also, we observed an inverse association of the IFN pathway expression and the susceptibility to NDV. The downregulated genes in NDV-GFP-sensitive cells were functionally enriched for antiviral mechanisms by interferon and immune system pathways, demonstrating that these mechanisms are the most prominent for oncolysis by NDV. To our knowledge, this is the first description of oncolysis by an NDV strain in canine mammary cancer cells. We also demonstrated specific molecular pathways related to NDV susceptibility in these cancer cells, opening the possibility to use NDV as a therapeutic-targeted option for more malignant CMCs. Therefore, these results urge for more studies using oncolytic NDVs, especially considering genetic editing to improve efficacy in dogs.


Asunto(s)
Enfermedades de los Perros , Neoplasias Mamarias Animales/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Antivirales , Enfermedades de los Perros/terapia , Perros , Femenino , Interferones , Virus de la Enfermedad de Newcastle , Viroterapia Oncolítica/veterinaria , Replicación Viral
12.
Front Immunol ; 11: 576658, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193370

RESUMEN

Immune evasion is an important cancer hallmark and the understanding of its mechanisms has generated successful therapeutic approaches. Induction of immunogenic cell death (ICD) is expected to attract immune cell populations that promote innate and adaptive immune responses. Here, we present a critical advance for our adenovirus-mediated gene therapy approach, where the combined p14ARF and human interferon-ß (IFNß) gene transfer to human melanoma cells led to oncolysis, ICD and subsequent activation of immune cells. Our results indicate that IFNß alone or in combination with p14ARF was able to induce massive cell death in the human melanoma cell line SK-MEL-147, though caspase 3/7 activation was not essential. In situ gene therapy of s.c. SK-MEL-147 tumors in Nod-Scid mice revealed inhibition of tumor growth and increased survival in response to IFNß alone or in combination with p14ARF. Emission of critical markers of ICD (exposition of calreticulin, secretion of ATP and IFNß) was stronger when cells were treated with combined p14ARF and IFNß gene transfer. Co-culture of previously transduced SK-MEL-147 cells with monocyte-derived dendritic cells (Mo-DCs) derived from healthy donors resulted in increased levels of activation markers HLA-DR, CD80, and CD86. Activated Mo-DCs were able to prime autologous and allogeneic T cells, resulting in increased secretion of IFNγ, TNF-α, and IL-10. Preliminary data showed that T cells primed by Mo-DCs activated with p14ARF+IFNß-transduced SK-MEL-147 cells were able to induce the loss of viability of fresh non-transduced SK-MEL-147 cells, suggesting the induction of a specific cytotoxic population that recognized and killed SK-MEL-147 cells. Collectively, our results indicate that p14ARF and IFNß delivered by our adenoviral system induced oncolysis in human melanoma cells accompanied by adaptive immune response activation and regulation.


Asunto(s)
Adenoviridae/fisiología , Inmunoterapia/métodos , Interferón beta/genética , Melanoma/terapia , Linfocitos T/inmunología , Proteína p14ARF Supresora de Tumor/genética , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Terapia Genética , Humanos , Activación de Linfocitos , Melanoma/genética , Ratones , Ratones SCID , Viroterapia Oncolítica , Carga Tumoral , Escape del Tumor
13.
Biomedica ; 40(2): 362-381, 2020 06 15.
Artículo en Inglés, Español | MEDLINE | ID: mdl-32673463

RESUMEN

INTRODUCTION: Cancer is the second leading cause of death in the United States, surpassed only by cardiovascular disease. However, cancer has now overtaken cardiovascular disease as the main cause of death in 12 countries in Western Europe. The burden of cancer is posing a major challenge to health care systems worldwide and demanding improvements in methods for cancer prevention, diagnosis, and treatment. Alternative and complementary strategies for orthodox surgery, radiotherapy, and chemotherapy need to be developed. OBJECTIVE: To determine the oncolytic potential of tumor cell-adapted rotavirus in terms of their ability to infect and lysate murine myeloma Sp2/0-Ag14 cells. MATERIALS AND METHODS: We inoculated rotaviruses Wt1-5, WWM, TRUYO, ECwt-O, and WTEW in Sp2/0-Ag14 cells and we examined their infectious effects by immunocytochemistry, immunofluorescence, flow cytometry, and DNA fragmentation assays. RESULTS: Rotavirus infection involved the participation of some heat shock proteins, of protein disulfide isomerase (PDI), and integrin ß3. We detected the accumulation of viral antigens within the virus-inoculated cells and in the culture medium in all the rotavirus isolates examined. The rotavirus-induced cell death mechanism in Sp2/0-Ag14 cells involved changes in cell membrane permeability, chromatin condensation, and DNA fragmentation, which were compatible with cytotoxicity and apoptosis. CONCLUSIONS: The ability of the rotavirus isolates Wt1-5, WWM, TRUYO, ECwt-O, and WTEW to infect and cause cell death of Sp2/0-Ag14 cells through mechanisms that are compatible with virus-induced apoptosis makes them potential candidates as oncolytic agents.


Introducción. El cáncer es la segunda causa de muerte en los Estados Unidos, solamente superado por la enfermedad cardiovascular. Sin embargo, el cáncer aventaja a la enfermedad cardiovascular como primera causa de muerte en doce países de Europa occidental. Se requieren mejores métodos de prevención, diagnóstico y tratamiento para afrontar el gran desafío que el cáncer representa mundialmente para los sistemas de salud, y se necesita desarrollar estrategias alternativas y complementarias a la cirugía, la radioterapia y la quimioterapia convencionales. Objetivo. Evaluar el potencial oncolítico de rotavirus adaptados a células tumorales por su capacidad para infectar y lisar células Sp2/0-Ag14 de mieloma de ratón. Materiales and métodos. Los aislamientos de rotavirus Wt1-5, WWM, TRUYO, ECwt-O y WTEW se inocularon en células Sp2/0-Ag14 y se examinaron sus efectos infecciosos mediante inmunocitoquímica, inmunofluorescencia, citometría de flujo y ensayos de fragmentación del ADN. Resultados. La infección con los rotavirus Wt1-5, WWM, TRUYO, ECwt-O y WTEW implicó la participación de algunas proteínas de choque térmico, la proteína disulfuro isomerasa y la integrina ß3. La acumulación de antígenos virales intracelulares y extracelulares se detectó en todos los virus utilizados. Los mecanismos de muerte inducidos por los rotavirus en células Sp2/0-Ag14 indujeron cambios en la permeabilidad de la membrana celular, la condensación de cromatina y la fragmentación de ADN, los cuales fueron compatibles con citotoxicidad y apoptosis. Conclusiones. La capacidad de los rotavirus estudiados para infectar y causar la muerte de células Sp2/0-Ag14 mediante mecanismos compatibles con la apoptosis inducida viralmente los convierte en candidatos potenciales para ser utilizados como agentes oncolíticos.


Asunto(s)
Efecto Citopatogénico Viral , Mieloma Múltiple/terapia , Viroterapia Oncolítica , Rotavirus , Secuencia de Aminoácidos , Animales , Antígenos Virales/análisis , Apoptosis , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Cromatina/ultraestructura , Medios de Cultivo/química , Fragmentación del ADN , Proteínas de Choque Térmico/fisiología , Integrina beta3/fisiología , Ratones , Mieloma Múltiple/patología , Proteína Disulfuro Isomerasas/fisiología , Rotavirus/inmunología , Rotavirus/fisiología , Replicación Viral
14.
Mediators Inflamm ; 2020: 8680692, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32410869

RESUMEN

Oncolytic virus therapy has been tested against cancer in preclinical models and clinical assays. Current evidence shows that viruses induce cytopathic effects associated with fusogenic protein-mediated syncytium formation and immunogenic cell death of eukaryotic cells. We have previously demonstrated that tumor cell bodies generated from cells expressing the fusogenic protein of the infectious salmon anemia virus (ISAV-F) enhance crosspriming and display prophylactic antitumor activity against melanoma tumors. In this work, we evaluated the effects of the expression of ISAV-F on the B16 melanoma model, both in vitro and in vivo, using chitosan nanoparticles as transfection vehicle. We confirmed that the transfection of B16 tumor cells with chitosan nanoparticles (NP-ISAV) allows the expression of a fusogenically active ISAV-F protein and decreases cell viability because of syncytium formation in vitro. However, the in vivo transfection induces a delay in tumor growth, without inducing changes on the lymphoid populations in the tumor and the spleen. Altogether, our observations show that expression of ISAV fusion protein using chitosan nanoparticles induces cell fusion in melanoma cells and slight antitumor response.


Asunto(s)
Antineoplásicos/farmacología , Quitosano/química , Melanoma/tratamiento farmacológico , Nanopartículas/química , Viroterapia Oncolítica/métodos , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Supervivencia Celular , Quitosano/metabolismo , ADN Complementario/metabolismo , Células Gigantes/metabolismo , Humanos , Isavirus/genética , Linfocitos/citología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nanomedicina/métodos , Infecciones por Orthomyxoviridae/genética , Proteínas Recombinantes de Fusión/química , Propiedades de Superficie , Transfección
15.
Mol Ther ; 28(5): 1276-1286, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32220305

RESUMEN

Malignant brain tumors are among the most aggressive cancers with poor prognosis and no effective treatment. Recently, we reported the oncolytic potential of Zika virus infecting and destroying the human central nervous system (CNS) tumors in vitro and in immunodeficient mice model. However, translating this approach to humans requires pre-clinical trials in another immunocompetent animal model. Here, we analyzed the safety of Brazilian Zika virus (ZIKVBR) intrathecal injections in three dogs bearing spontaneous CNS tumors aiming an anti-tumoral therapy. We further assessed some aspects of the innate immune and inflammatory response that triggers the anti-tumoral response observed during the ZIKVBR administration in vivo and in vitro. For the first time, we showed that there were no negative clinical side effects following ZIKVBR CNS injections in dogs, confirming the safety of the procedure. Furthermore, the intrathecal ZIKVBR injections reduced tumor size in immunocompetent dogs bearing spontaneous intracranial tumors, improved their neurological clinical symptoms significantly, and extended their survival by inducing the destruction specifically of tumor cells, sparing normal neurons, and activating an immune response. These results open new perspectives for upcoming virotherapy using ZIKV to destroy and induce an anti-tumoral immune response in CNS tumors for which there are currently no effective treatments.


Asunto(s)
Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/terapia , Viroterapia Oncolítica/métodos , Seguridad del Paciente , Carga Tumoral , Infección por el Virus Zika/complicaciones , Virus Zika/inmunología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perros , Inmunidad , Inyecciones Espinales , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/virología , Monocitos/inmunología , Monocitos/virología , Neuronas/metabolismo , Neuronas/virología , Resultado del Tratamiento
16.
Artículo en Portugués | VETINDEX | ID: biblio-1489067

RESUMEN

O câncer continua a ser uma das principais causas de mortes em humanos e em cães. Os tumores de ocorrência natural em cães têm muitas similaridades biológicas com os cânceres humanos e respondem similarmente às terapias convencionais. Por isso, pode-se inferir que os protocolos clínicos adotados para os humanos poderiam ser transferidos para o tratamento de cães com câncer e que terapêuticas com êxito no tratamento de tais animais poderiam servir para o tratamento de seres humanos. A viroterapia oncolítica em cães é o objeto deste artigo de revisão.


Cancer remains a leading cause of death in humans and dogs. Naturally occurring tumors in dogs have many biological similarities to human cancers, and respond similarly to conventional therapies. Thus, it can be inferred that human clinical protocols could be transferable for the treatment of dogs with cancer and that successful therapies in the treatment of these animals could be used for the treatment of human beings. Oncolytic virotherapy in dogs is the subject of this review article.


Asunto(s)
Animales , Perros , Perros , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Viroterapia Oncolítica/veterinaria , Virus Oncolíticos
17.
R. Educ. contin. Med. Vet. Zoot. ; 18(3): e38125, 2020.
Artículo en Portugués | VETINDEX | ID: vti-31028

RESUMEN

O câncer continua a ser uma das principais causas de mortes em humanos e em cães. Os tumores de ocorrência natural em cães têm muitas similaridades biológicas com os cânceres humanos e respondem similarmente às terapias convencionais. Por isso, pode-se inferir que os protocolos clínicos adotados para os humanos poderiam ser transferidos para o tratamento de cães com câncer e que terapêuticas com êxito no tratamento de tais animais poderiam servir para o tratamento de seres humanos. A viroterapia oncolítica em cães é o objeto deste artigo de revisão.(AU)


Cancer remains a leading cause of death in humans and dogs. Naturally occurring tumors in dogs have many biological similarities to human cancers, and respond similarly to conventional therapies. Thus, it can be inferred that human clinical protocols could be transferable for the treatment of dogs with cancer and that successful therapies in the treatment of these animals could be used for the treatment of human beings. Oncolytic virotherapy in dogs is the subject of this review article.(AU)


Asunto(s)
Animales , Perros , Perros , Viroterapia Oncolítica/métodos , Viroterapia Oncolítica/veterinaria , Neoplasias/terapia , Virus Oncolíticos
18.
Curr Oncol Rep ; 21(10): 94, 2019 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-31606796

RESUMEN

PURPOSE OF REVIEW: Recurrent glioblastoma (rGBM) has no standard treatment. Despite a better molecular knowledge, few therapies have brought changes in clinical practice so far. Here we will review the current data evaluating the re-radiation, re-resection, bevacizumab, and cytotoxic chemotherapy agents in this setting. We will also discuss the advances of immunotherapy and the possible benefit of this treatment for patients with rGBM. RECENT FINDINGS: Next-generation sequencing is increasingly utilized in the clinical practice of neuro-oncologists, bringing gene mutations as targets for therapies. As in other solid tumors, immunotherapy has been also extensively studied in rGBM, with interesting results in phase I and II trials. The most promising therapies in the horizon are combinations including immune checkpoint inhibitors, virotherapy, vaccines, and monoclonal antibodies. Although re-radiation, re-resection, bevacizumab, and chemotherapy are still the most widely used therapies for treating rGBM, the clinical benefit from these treatments is still not well established. Preliminary results of studies with immune checkpoint inhibitors were disappointing, but virotherapy emerges as more promising immunotherapy in rGBM, especially in combination with other strategies. In addition to the gain in overall survival, the improvement in the quality of life of these patients is also expected.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Glioblastoma/terapia , Viroterapia Oncolítica/métodos , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/virología , Ensayos Clínicos como Asunto , Terapia Combinada , Glioblastoma/inmunología , Glioblastoma/patología , Glioblastoma/virología , Humanos , Pronóstico , Ensayos Clínicos Controlados Aleatorios como Asunto
19.
Cancer Res ; 78(12): 3363-3374, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29700002

RESUMEN

Zika virus (ZIKV) is largely known for causing brain abnormalities due to its ability to infect neural progenitor stem cells during early development. Here, we show that ZIKV is also capable of infecting and destroying stem-like cancer cells from aggressive human embryonal tumors of the central nervous system (CNS). When evaluating the oncolytic properties of Brazilian Zika virus strain (ZIKVBR) against human breast, prostate, colorectal, and embryonal CNS tumor cell lines, we verified a selective infection of CNS tumor cells followed by massive tumor cell death. ZIKVBR was more efficient in destroying embryonal CNS tumorspheres than normal stem cell neurospheres. A single intracerebroventricular injection of ZIKVBR in BALB/c nude mice bearing orthotopic human embryonal CNS tumor xenografts resulted in a significantly longer survival, decreased tumor burden, fewer metastasis, and complete remission in some animals. Tumor cells closely resembling neural stem cells at the molecular level with activated Wnt signaling were more susceptible to the oncolytic effects of ZIKVBR Furthermore, modulation of Wnt signaling pathway significantly affected ZIKVBR-induced tumor cell death and viral shedding. Altogether, these preclinical findings indicate that ZIKVBR could be an efficient agent to treat aggressive forms of embryonal CNS tumors and could provide mechanistic insights regarding its oncolytic effects.Significance: Brazilian Zika virus strain kills aggressive metastatic forms of human CNS tumors and could be a potential oncolytic agent for cancer therapy. Cancer Res; 78(12); 3363-74. ©2018 AACR.


Asunto(s)
Neoplasias del Sistema Nervioso Central/terapia , Neoplasias de Células Germinales y Embrionarias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Virus Zika/fisiología , Animales , Encéfalo/citología , Neoplasias del Sistema Nervioso Central/mortalidad , Neoplasias del Sistema Nervioso Central/patología , Humanos , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de Células Germinales y Embrionarias/mortalidad , Neoplasias de Células Germinales y Embrionarias/patología , Células-Madre Neurales/patología , Análisis de Supervivencia , Resultado del Tratamiento , Esparcimiento de Virus , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Methods Mol Biol ; 1651: 113-130, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28801903

RESUMEN

Here we describe a collection of methods that have been adapted to isolate and modify tumor-specific promoters (TSPs ) to drive viral replication for cancer therapy and other uses. We will describe as examples the secreted protein acidic and rich in cysteine (SPARC ) and the protease-activated receptor-1 (PAR-1) promoter. We outline strategies to select appropriate TSPs using bioinformatics resources and the methods utilized in their subsequent cloning, assessment of transcriptional activity, and their use in conditionally replicative oncolytic adenoviruses .


Asunto(s)
Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Osteonectina/genética , Regiones Promotoras Genéticas , Receptor PAR-1/genética , Activación Transcripcional , Replicación Viral , Adenoviridae/genética , Animales , Clonación Molecular/métodos , Regulación Viral de la Expresión Génica , Vectores Genéticos/genética , Genómica/métodos , Humanos , Ratones , Neoplasias/genética , Neoplasias/terapia , Plásmidos/genética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA