Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vet Comp Oncol ; 21(3): 520-532, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37282822

RESUMO

Spontaneous canine mammary carcinomas (CMCs) have been widely considered a good research model for human breast cancers, which brings much attention to CMCs. In recent years, the oncolytic effect of Newcastle disease virus (NDV) on cancer cells has been widely studied, but its effect on CMCs is still unclear. This study aims to investigate the oncolytic effect of NDV LaSota strain on canine mammary carcinoma cell line (CMT-U27) in vivo and in vitro. The in vitro cytotoxicity and immunocytochemistry experiments showed that NDV selectively replicated in CMT-U27 cells, and inhibited cell proliferation and migration but not in MDCK cells. KEGG analysis of transcriptome sequencing indicated the importance of the TNFα and NF-κB signalling pathways in the anti-tumour effect of NDV. Subsequently, the significantly increased expression of TNFα, p65, phospho-p65, caspase-8, caspase-3 and cleaved-PARP proteins in the NDV group suggested that NDV induced CMT-U27 cells apoptosis by activating the caspase-8/caspase-3 pathway and the TNFα/NF-κB signalling pathway. Nude mice tumour-bearing experiments showed that NDV could significantly decrease the growth rate of CMC in vivo. In conclusion, our study demonstrates the effective oncolytic effects of NDV on CMT-U27 cells in vivo and in vitro, and suggests NDV as a promising candidate for oncolytic therapy.


Assuntos
Carcinoma , Doenças do Cão , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Cães , Camundongos , Apoptose , Carcinoma/veterinária , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Doenças do Cão/terapia , Camundongos Nus , Vírus da Doença de Newcastle/metabolismo , NF-kappa B/metabolismo , Terapia Viral Oncolítica/veterinária , Fator de Necrose Tumoral alfa/metabolismo
2.
Vet Med Sci ; 9(3): 1426-1437, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36920334

RESUMO

BACKGROUND: Breast cancer is one of the most common malignancies in women, with one in 20 globally. Oncolytic viruses have recently been the first step in the biological treatment of cancer, either genetically engineered or naturally occurring. They increase specifically inside cancer cells and destroy them without damaging normal tissues or producing a host immune response against tumour cells or expressing transgenes. One of the most known members of this family is the Newcastle disease virus (NDV), a natural oncolytic virus that selectively induces apoptosis and DNA fragmentation in human cancer cells. METHODS: This study performed biochemical and molecular investigations with variable doses of NDV (32, 64, 128 HAU) and liposomal doxorubicin (9 mg/kg) on mouse triple-negative mammary carcinoma cell line 4T1 and BALB/c models tumours for the first time. RESULTS: Real-time quantitative PCR analysis in NDV-treated animal tumours showed increased expression of P21, P27 and P53 genes and decreased expression of CD34, integrin Alpha 5, VEGF and VEGF-R genes. Additional assessments in treated mouse models also showed that NDV increased ROS production, induced apoptosis, reduced tumour size and significantly improved prognosis, with no adverse effect on normal tissues. CONCLUSIONS: These findings all together might indicate that NDV in combination with chemotherapy drugs could improve prognosis in cancer patients although many more conditions should be considered.


Assuntos
Neoplasias da Mama , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Feminino , Camundongos , Animais , Vírus Oncolíticos/genética , Vírus da Doença de Newcastle/genética , Terapia Viral Oncolítica/veterinária , Fator A de Crescimento do Endotélio Vascular , Linhagem Celular , Neoplasias da Mama/veterinária
3.
Res Vet Sci ; 139: 159-165, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34332418

RESUMO

Recombinant Newcastle disease virus vectors have gained a lot of interest for its oncolytic virus therapy and cancer immune therapeutic properties due to its selective replication to high titers in cancer cells. The aim of this study was to find out the oncolytic effects of mesogenic recombinant NDV strain R2B-GFP on murine mammary tumor cell line 4T1 and murine melanoma cell line B16-F10. The anti-tumor effects of R2B-GFP virus were studied via expression of virus transgene GFP in cancer cells, evaluating its cytotoxicity and cell migration efficacies by MTT and wound healing assays respectively. In addition, the underlying apoptotic mechanism of R2B-GFP virus was estimated by TUNEL assay, colorimetric estimation of Caspase-3, 8 and 9 and the estimation of Bax to Bcl-2 ratio. The results showed a significant decrease in viability of both 4T1 and B16-F10 cells infected with R2B-GFP virus at 0.1 and 1 MOI. R2B-GFP virus could significantly induce apoptosis in the 4T1 and B16-F10 cells as compared to the uninfected control. Further, a flow cytometry analysis on apoptotic cells percentage and mitochondria membrane permeability test was also studied in R2B-GFP virus treated 4T1 and B16-F10 cell lines. The R2B-GFP virus caused an increase in loss of mitochondrial membrane permeability in both 4T1 and B16-F10 cells indicating the involvement of mitochondrial regulated cell death. Thus, the recombinant virus R2B-GFP virus proved to be a valid candidate for oncolytic viral therapy in 4T1 and B16-F10 cells.


Assuntos
Neoplasias Mamárias Animais , Melanoma , Vírus da Doença de Newcastle , Terapia Viral Oncolítica , Doenças dos Roedores , Animais , Apoptose , Linhagem Celular Tumoral , Neoplasias Mamárias Animais/terapia , Melanoma/terapia , Melanoma/veterinária , Camundongos , Vírus da Doença de Newcastle/patogenicidade , Terapia Viral Oncolítica/veterinária
4.
Vet Comp Oncol ; 19(3): 593-601, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33871162

RESUMO

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.


Assuntos
Doenças do Cão , Neoplasias Mamárias Animais/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Antivirais , Doenças do Cão/terapia , Cães , Feminino , Interferons , Vírus da Doença de Newcastle , Terapia Viral Oncolítica/veterinária , Replicação Viral
5.
Res Vet Sci ; 133: 313-317, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33183781

RESUMO

The prognosis of canine transitional cell carcinoma (TCC) of urinary bladder is generally poor because it is difficult to diagnose at early stages and conventional therapies, such as surgical resection and/or chemotherapy, are often not curative treatments. Based on our previous report that recombinant measles virus (rMV-SLAMblind) therapy could be a new treatment for canine mammary tumor, the applicability of rMV-SLAMblind in canine urinary bladder TCC was examined in this study. A canine TCC cell line was established from a TCC patient dog by transplanting a piece of the tumor mass into an immunodeficient mouse and then isolating the primary TCC cells from the grown tumor mass. The primary cultured cells, named TCC-NU1, express nectin-4, a receptor for rMV-SLAMblind infection. The rMV-SLAMblind infected TCC-NU1 cells, and dose-dependently showed cell cytotoxicity. Moreover, intratumoral administration of rMV-SLAMblind in a xenograft model bearing TCC-NU1 cells significantly suppressed the tumor growth reducing the endpoint mass of tumors in treated mice compared to control mice. These results suggest that virotherapy with rMV-SLAMblind be a new candidate therapy for canine TCC.


Assuntos
Carcinoma de Células de Transição/terapia , Doenças do Cão/terapia , Vírus do Sarampo/fisiologia , Terapia Viral Oncolítica/veterinária , Neoplasias da Bexiga Urinária/veterinária , Animais , Carcinoma de Células de Transição/veterinária , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Cães , Feminino , Humanos , Camundongos , Vírus Oncolíticos/metabolismo , Neoplasias da Bexiga Urinária/terapia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Vet Comp Oncol ; 18(1): 36-42, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31618515

RESUMO

Replicating oncolytic viruses (OVs) are appealing, new, FDA-approved, therapeutic options for humans with head and neck cancers and melanomas. These treatments are not yet available for veterinary patients, but recent clinical trials have shown several OVs to be safe in dogs and cats. Specific viruses being used to treat sarcomas in dogs include modified canine adenovirus 2, myxoma virus, vesicular stomatitis virus and reovirus. In cats with vaccine-associated sarcomas, poxviruses have been injected postoperatively and a reduced rate of tumour recurrence was documented. To date, the response rates of canine and feline patients to OV therapy have been variable (as they are in people). Optimal methods of OV administration and dosing schedules continue to be evaluated. One way to improve outcomes of OV therapy in veterinary patients may be to use OVs in combination with other immunomodulatory therapies. This review discusses the potential utility of concurrent therapy with an OV and an inhibitor of the type I interferon pathway.


Assuntos
Antineoplásicos/uso terapêutico , Doenças do Gato/tratamento farmacológico , Doenças do Cão/tratamento farmacológico , Interferon Tipo I/uso terapêutico , Terapia Viral Oncolítica/veterinária , Sarcoma/veterinária , Neoplasias de Tecidos Moles/veterinária , Animais , Gatos , Cães , Terapia Viral Oncolítica/métodos , Sarcoma/tratamento farmacológico , Sarcoma/patologia , Neoplasias de Tecidos Moles/tratamento farmacológico , Neoplasias de Tecidos Moles/patologia
7.
Vet Comp Oncol ; 17(2): 184-193, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30761736

RESUMO

Canine histiocytic sarcoma is an aggressive, fatal neoplastic disease with a poor prognosis. Lomustine is generally accepted as the first-line systemic therapy, although this compound does not provide complete regression. Therefore, research into a novel approach against canine histiocytic sarcoma is needed. However, anti-tumour effects of oncolytic therapy using reovirus against histiocytic sarcoma are unknown. Here, we showed that reovirus has oncolytic activity in canine histiocytic sarcoma cell lines in vitro and in vivo. We found that reovirus can replicate and induce caspase-dependent apoptosis in canine histiocytic sarcoma cell lines. A single intra-tumoural injection of reovirus completely suppressed the growth of subcutaneously grafted tumours in NOD/SCID mice. Additionally, we demonstrated that susceptibility to reovirus-induced cell death was attributable to the extent of expression of type I interferons induced by reovirus infection in vitro. In conclusion, oncolytic reovirus appears to be an effective treatment option for histiocytic sarcoma, and therefore warrants further investigation in early clinical trials.


Assuntos
Doenças do Cão/virologia , Sarcoma Histiocítico/veterinária , Terapia Viral Oncolítica/veterinária , Vírus Oncolíticos/patogenicidade , Orthoreovirus de Mamíferos/patogenicidade , Animais , Morte Celular , Linhagem Celular Tumoral/virologia , Cães , Sarcoma Histiocítico/virologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Terapia Viral Oncolítica/métodos , Reação em Cadeia da Polimerase em Tempo Real/veterinária
8.
Vet Comp Oncol ; 16(2): 229-238, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29076241

RESUMO

Oncolytic virotherapy is a novel treatment involving replication-competent virus in the elimination of cancer. We have previously reported the oncolytic effects of reovirus in various canine cancer cell lines. This study aims to establish the safety profile of reovirus in dogs with spontaneously occurring tumours and to determine a recommended dosing regimen. Nineteen dogs with various tumours, mostly of advanced stages, were treated with reovirus, ranging from 1.0 × 108 to 5.0 × 109 TCID50 given as intratumour injection (IT) or intravenous infusion (IV) daily for up to 5 consecutive days in 1 or multiple treatment cycles. Adverse events (AEs) were graded according to the Veterinary Cooperative Oncology Group- Common Terminology Criteria for Adverse Events (VCOG-CTCAE) v1.1 guidelines. Viral shedding, neutralizing anti-reovirus antibody (NARA) production and immunohistochemical (IHC) detection of reovirus protein in the tumours were also assessed. AE was not observed in most dogs and events were limited to Grade I or II fever, vomiting, diarrhoea and inflammation of the injected tumour. No infectious virus was shed and all dogs had elevated NARA levels post-treatment. Although IHC results were only available in 6 dogs, 4 were detected positive for reovirus protein. In conclusion, reovirus is well-tolerated and can be given safely to tumour-bearing dogs according to the dosing regimen used in this study without significant concerns of viral shedding. Reovirus is also potentially effective in various types of canine tumours.


Assuntos
Doenças do Cão/tratamento farmacológico , Doenças do Cão/imunologia , Neoplasias/veterinária , Terapia Viral Oncolítica/veterinária , Vírus Oncolíticos/imunologia , Reoviridae/imunologia , Animais , Anticorpos Neutralizantes/sangue , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Cães , Feminino , Japão , Masculino , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Terapia Viral Oncolítica/métodos , Projetos Piloto , Reação em Cadeia da Polimerase , Faculdades de Medicina Veterinária , Resultado do Tratamento , Eliminação de Partículas Virais
9.
Vet Comp Oncol ; 15(2): 336-344, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26373887

RESUMO

Oncolytic virotherapy is a novel strategy for treatment of cancer in humans and companion animals as well. Canine distemper virus (CDV), a paramyxovirus, has proven to be oncolytic through induction of apoptosis in canine-derived tumour cells, yet the mechanism behind this inhibitory action is poorly understood. In this study, three human mammary tumour cell lines and one canine-derived adenofibrosarcoma cell line were tested regarding to their susceptibility to CDV infection, cell proliferation, apoptosis, mitochondrial membrane potential and expression of tumour necrosis factor-alpha-induced protein 8 (TNFAIP8). CDV replication-induced cytopathic effect, decrease of cell proliferation rates, and >45% of infected cells were considered death and/or under late apoptosis/necrosis. TNFAIP8 and CDVM gene expression were positively correlated in all cell lines. In addition, mitochondrial membrane depolarization was associated with increase in virus titres (p < 0.005). Thus, these results strongly suggest that both human and canine mammary tumour cells are potential candidates for studies concerning CDV-induced cancer therapy.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Vírus da Cinomose Canina/metabolismo , Terapia Viral Oncolítica/veterinária , Adenocarcinoma/terapia , Adenocarcinoma/veterinária , Animais , Apoptose , Neoplasias da Mama/terapia , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células , Doenças do Cão/terapia , Cães , Feminino , Humanos , Neoplasias Mamárias Animais/terapia , Terapia Viral Oncolítica/métodos
10.
Vet Comp Oncol ; 14 Suppl 1: 61-73, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25319493

RESUMO

Reovirus is a potent oncolytic virus in many human neoplasms that has reached phase II and III clinical trials. Our laboratory has previously reported the oncolytic effects of reovirus in canine mast cell tumour (MCT). In order to further explore the potential of reovirus in veterinary oncology, we tested the susceptibility of reovirus in 10 canine lymphoma cell lines. Reovirus-induced cell death, virus replication and infectivity were confirmed in four cell lines with variable levels of susceptibility. The level of Ras activation varied among the cell lines with no correlation with reovirus susceptibility. Reovirus-susceptible cell lines underwent apoptosis as proven by propidium iodide (PI) staining, Annexin V-FITC/PI assay, cleavage of PARP and inhibition of cell death by caspase inhibitor. A single intratumoral injection of reovirus suppressed the growth of canine lymphoma subcutaneous tumour in NOD/SCID mice. Unlike canine MCT, canine lymphoma is less susceptible to reovirus.


Assuntos
Doenças do Cão/patologia , Doenças do Cão/virologia , Linfoma não Hodgkin/veterinária , Terapia Viral Oncolítica/veterinária , Reoviridae/fisiologia , Animais , Western Blotting/veterinária , Morte Celular , Linhagem Celular Tumoral/virologia , Cães , Linfoma não Hodgkin/patologia , Linfoma não Hodgkin/virologia , Terapia Viral Oncolítica/métodos
11.
Vet Comp Oncol ; 7(3): 153-61, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19691644

RESUMO

The use of viruses to treat cancer has been studied for decades. With the advancement of molecular biology, viruses have been modified and genetically engineered to optimize their ability to target cancer cells. Canine viruses, such as distemper virus and adenovirus, are being exploited for the treatment of canine cancer as the dog has proven to be a good comparative model for human cancer research and proof of concept investigations. In this review, we introduce the concept of oncolytic viruses and describe some of the preliminary attempts to use oncolytic viruses for the treatment of canine cancer.


Assuntos
Doenças do Cão/terapia , Terapia Genética/veterinária , Neoplasias/veterinária , Terapia Viral Oncolítica/veterinária , Vírus/classificação , Animais , Doenças do Cão/virologia , Cães , Engenharia Genética , Terapia Genética/métodos , Neoplasias/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...