Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Oncolytics ; 24: 864-872, 2022 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-35317522

RESUMO

Pancreatic cancer resistance to immunotherapies is partly due to deficits in tumor-infiltrating immune cells and stromal density. Combination therapies that modify stroma and recruit immune cells are needed. Vitamin D analogs such as calcipotriol (Cal) decrease fibrosis in pancreas stroma, thus allowing increased chemotherapy delivery. OVs infect, replicate in, and kill cancer cells and recruit immune cells to immunodeficient microenvironments. We investigated whether stromal modification with Cal would enhance oncolytic viroimmunotherapy using recombinant orthopoxvirus, CF33. We assessed effect of Cal on CF33 replication using pancreas ductal adenocarcinoma (PDAC) cell lines and in vivo flank orthotopic models. Proliferation assays showed that Cal did not alter viral replication. Less replication was seen in cell lines whose division was slowed by Cal, but this appeared proportional to cell proliferation. Three-dimensional in vitro models demonstrated decreased myofibroblast integrity after Cal treatment. Cal increased vascular lumen size and immune cell infiltration in subcutaneous models of PDAC and increased viral delivery and replication. Cal plus serial OV dosing in the syngeneic Pan02 model caused more significant tumor abrogation than other treatments. Cal-treated tumors had less dense fibrosis, enhanced immune cell infiltration, and decreased T cell exhaustion. Calcipotriol is a possible adjunct for CF33-based oncolytic viroimmunotherapy against PDAC.

2.
Mol Ther Oncolytics ; 23: 303-310, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34786474

RESUMO

Oncolytic viruses infect, replicate in, and kill cancer cells, leaving normal cells unharmed; they also recruit and activate immune cells against tumor cells. While clinical indications for viroimmunotherapy are growing, barriers to widespread treatment remain. Ensuring real-time tracking of viral replication and resulting anti-tumor immune responses will overcome some of these barriers and is thus a top priority. Clinically optimizing trackability of viral replication will promote safe dose increases, guide serial dosing, and enhance treatment effects. However, viral delivery is only half the story. Oncolytic viruses are known to upregulate immune checkpoint expression, thereby priming otherwise immunodeficient tumor immune microenvironments for treatment with checkpoint inhibitors. Novel modalities to track virus-induced changes in tumor microenvironments include non-invasive measurements of immune cell populations and responses to viroimmunotherapy such as (1) in situ use of radiotracers to track checkpoint protein expression or immune cell traffic, and (2) ex vivo labeling of immune cells followed by nuclear medicine imaging. Herein, we review clinical progress toward accurate imaging of oncolytic virus replication, and we further review the current status of functional imaging of immune responses to viroimmunotherapy.

3.
Mol Cancer Ther ; 20(1): 173-182, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33262221

RESUMO

Although it is known that oncolytic viruses can inflame and recruit immune cells to otherwise immunosuppressed tumor microenvironments, the influence of the antiviral immune response on antitumor immunity is less clear across viral platforms and tumor types. CF33 is a recombinant orthopoxvirus backbone effective against colon cancer. We tested derivatives of CF33 with and without immune-checkpoint inhibition (anti-PD-L1) in mouse models of colon cancer. Results showed that the efficacy of CF33 backbone with J2R deletion (single-deleted) against colon cancer is not altered by additional deletion of F14.5L in vitro or in vivo CF33 infection upregulated PD-L1 expression on tumor cells and led to an increased influx of lymphocytes and macrophages in tumors. Also, the levels of active CD8+ (IFNγ+) T cells in the virus-treated tumors were higher than those in control-treated tumors. Furthermore, a combination of CF33 derivatives with anti-PD-L1 resulted in durable tumor regression and long-term survival, resistant to tumor rechallenge. Analysis of immune cells from the treated mice showed that tumor-specific T cell activation occurred more robustly in tumors treated with the virus and that T cells were more strongly activated against the virus than against tumor, in an MHC-I-dependent manner. Our findings warrant further studies on the role of cross-priming of T cells against viral and tumor antigens, in the overall success of viroimmunotherapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/virologia , Apresentação Cruzada/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunidade , Orthopoxvirus/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular , Neoplasias do Colo/tratamento farmacológico , Apresentação Cruzada/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Memória Imunológica/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Receptor de Morte Celular Programada 1/metabolismo , Recombinação Genética/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia
4.
Int J Mol Sci ; 21(19)2020 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-33023064

RESUMO

Oncolytic viroimmunotherapy is an exciting modality that can offer lasting anti-tumor immunity for aggressive malignancies like colon cancer. The impact of oncolytic viruses may be extended by combining them with agents to prime a tumor for viral susceptibility. This study investigates vitamin D analogue as an adjunct to oncolytic viral therapy for colon cancer. While vitamin D (VD) has historically been viewed as anti-viral, our in vitro investigations using human colon cancer cell lines showed that VD does not directly inhibit replication of recombinant chimeric poxvirus CF33. VD did restrict growth in HT29 but not HCT116 human colon cancer cells. In vivo investigations using HCT116 and HT29 xenograft models of colon cancer demonstrated that a VD analogue, calcipotriol, was additive with CF33-based viral therapy in VD-responsive HT29 but not in HCT116 tumors. Analyses of RNA-sequencing and gene expression data demonstrated a downregulation in the Jak-STAT signaling pathway with the addition of VD to viral therapy in HT29 models suggesting that the anti-inflammatory properties of VD may enhance the effects of viral therapy in some models. In conclusion, VD may prime oncolytic viral therapy in certain colon cancers.


Assuntos
Neoplasias do Colo/terapia , Terapia Viral Oncolítica , Replicação Viral/efeitos dos fármacos , Vitamina D/farmacologia , Animais , Sequência de Bases/genética , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/virologia , Terapia Combinada , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Células HT29 , Humanos , Imunoterapia/métodos , Camundongos , Vírus Oncolíticos/genética , Vitamina D/genética , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncoimmunology ; 9(1): 1729300, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32158622

RESUMO

Triple-negative breast cancer is the most aggressive subtype of breast cancer and is difficult to treat. Breast cancer is considered to be poorly immunogenic and hence is less responsive to immunotherapies. We tested whether the oncolytic poxvirus CF33-hNIS-ΔF14.5 could modulate tumor immune microenvironment and make the tumors responsive to the immune checkpoint inhibitor anti-PD-L1. We found that virus infection causes the upregulation of PD-L1 levels on triple-negative breast cancer cells in vitro as well as in vivo in mice. In a mouse model of orthotopic triple-negative breast cancer, the virus was found to increase tumor infiltration by CD8+ T cells. Likewise, in mice treated with CF33-hNIS-ΔF14.5 high levels of proinflammatory cytokines IFNγ and IL-6 were found in the tumors but not in the serum. The levels of immune modulation were even higher in mice that were treated with a combination of the virus and anti-PD-L1 antibody. While CF33-hNIS-ΔF14.5 and anti-PD-L1 antibody failed to exert significant anti-tumor effect as a single agent, a combination of the two agents resulted in significant anti-tumor effect with 50% mice experiencing complete tumor regression when both agents were injected intra-tumorally. Furthermore, the 'cured' mice did not develop tumor after re-challenge with the same cancer cells suggesting that they developed immunity against those cancer cells. Taken together, our study shows that CF33-hNIS-ΔF14.5 favorably modulates tumor immune microenvironment in triple-negative breast cancer model making them responsive to the immune checkpoint inhibitor anti-PD-L1, and hence warrants further studies to determine the clinical applicability of this combination therapy.


Assuntos
Antígeno B7-H1 , Imunoterapia , Terapia Viral Oncolítica , Neoplasias de Mama Triplo Negativas , Animais , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral , Humanos , Camundongos , Neoplasias de Mama Triplo Negativas/terapia , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...