Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 12(4)2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38580330

RESUMEN

BACKGROUND: Initiation of antitumor immunity is reliant on the stimulation of dendritic cells (DCs) to present tumor antigens to naïve T cells and generate effector T cells that can kill cancer cells. Induction of immunogenic cell death after certain types of cytotoxic anticancer therapies can stimulate T cell-mediated immunity. However, cytotoxic therapies simultaneously activate multiple types of cellular stress and programmed cell death; hence, it remains unknown what types of cancer cell death confer superior antitumor immunity. METHODS: Murine cancer cells were engineered to activate apoptotic or pyroptotic cell death after Dox-induced expression of procell death proteins. Cell-free supernatants were collected to measure secreted danger signals, cytokines, and chemokines. Tumors were formed by transplanting engineered tumor cells to specifically activate apoptosis or pyroptosis in established tumors and the magnitude of immune response measured by flow cytometry. Tumor growth was measured using calipers to estimate end point tumor volumes for Kaplan-Meier survival analysis. RESULTS: We demonstrated that, unlike apoptosis, pyroptosis induces an immunostimulatory secretome signature. In established tumors pyroptosis preferentially activated CD103+ and XCR1+ type I conventional DCs (cDC1) along with a higher magnitude and functionality of tumor-specific CD8+ T cells and reduced number of regulatory T cells within the tumor. Depletion of cDC1 or CD4+ and CD8+ T cells ablated the antitumor response leaving mice susceptible to a tumor rechallenge. CONCLUSION: Our study highlights that distinct types of cell death yield varying immunotherapeutic effect and selective activation of pyroptosis can be used to potentiate multiple aspects of the anticancer immunity cycle.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Ratones , Animales , Piroptosis , Células Dendríticas , Citocinas/metabolismo
2.
Cancer Immunol Res ; 12(1): 7-16, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-37902605

RESUMEN

Cytotoxic anticancer therapies activate programmed cell death in the context of underlying stress and inflammatory signaling to elicit the emission of danger signals, cytokines, and chemokines. In a concerted manner, these immunomodulatory secretomes stimulate antigen presentation and T cell-mediated anticancer immune responses. In some instances, cell death-associated secretomes attract immunosuppressive cells to promote tumor progression. As it stands, cancer cell death-induced changes in the tumor microenvironment that contribute to antitumor or protumor effects remain largely unknown. This is complicated to examine because cell death is often subverted by tumors to circumvent natural, and therapy-induced, immunosurveillance. Here, we provide insights into important but understudied aspects of assessing the contribution of cell death to tumor elimination or cancer progression, including the role of tumor-associated genetics, epigenetics, and oncogenic factors in subverting immunogenic cell death. This perspective will also provide insights on how future studies may address the complex antitumor and protumor immunologic effects of cell death, while accounting for variations in tumor genetics and underlying microenvironment.


Asunto(s)
Apoptosis , Neoplasias , Humanos , Neoplasias/etiología , Muerte Celular , Citocinas/metabolismo , Presentación de Antígeno , Microambiente Tumoral
3.
Vet Immunol Immunopathol ; 243: 110367, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34923192

RESUMEN

Urothelial carcinoma (UC) is the most common urinary tumor in dogs and despite combinational therapies, only modest improvements in survival have been achieved in recent years. Given the utility of monoclonal antibodies against PD-1 and PD-L1 in human UC, we evaluated the protein and mRNA expression in three established canine urothelial carcinoma cell lines. Flow cytometry and western blot analysis confirmed cell line expression of both molecules in varying degrees. Reverse transcription PCR (RT-PCR) documented mRNA expression in all three cell lines for both PD-1 and PD-L1. Fluorescence microscopy was consistent with strong PD-1 and PD-L1 expression in the canine cell lines and was in line with previous human literature. Importantly, the flow cytometry work described in this study revealed higher cell intrinsic PD-1 expression in these cell lines which may have implications for tumor behavior and potential treatment opportunities in the future. Further work is necessary to determine the expression patterns in canine UC and potential for benefit with immunotherapy directed against PD-1 and PD-L1.


Asunto(s)
Antígeno B7-H1 , Carcinoma de Células Transicionales , Receptor de Muerte Celular Programada 1/genética , Neoplasias de la Vejiga Urinaria , Animales , Antígeno B7-H1/genética , Carcinoma de Células Transicionales/veterinaria , Línea Celular Tumoral , Enfermedades de los Perros , Perros , ARN Mensajero , Neoplasias de la Vejiga Urinaria/veterinaria
4.
PLoS One ; 16(8): e0255591, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34352013

RESUMEN

BACKGROUND: Canine urothelial carcinoma is the most common form of canine bladder cancer. Treatment with chemotherapy has variable response rates leading to most dogs succumbing to their disease within a year. Cannabidiol is an emerging treatment within the field of oncology. In reported in vivo studies, cannabidiol has induced apoptosis, reduced cell migration, and acted as a chemotherapy sensitizer in various human tumor types. The aim of this study was to characterize the effects of cannabidiol on canine urothelial carcinoma cell viability and apoptosis as both a single agent and in combination with chemotherapy in vitro. RESULTS: Cannabidiol reduced cell viability and induced apoptosis in canine urothelial cells as determined by crystal violet viability assay and annexin V/propidium iodide flow cytometry. Furthermore, combinations of cannabidiol with mitoxantrone and vinblastine chemotherapy yielded significantly reduced cell viability and increased apoptosis compared to single agent treatment alone. The drug interactions were deemed synergistic based on combination index calculations. Conversely, the combination of cannabidiol and carboplatin did not result in decreased cell viability and increased apoptosis compared to single agent treatment. Combination index calculations suggested an antagonistic interaction between these drugs. Finally, the combination of the non-steroidal anti-inflammatory drug piroxicam with cannabidiol did not significantly affect cell viability, although, some cell lines demonstrated decreased cell viability when mitoxantrone was combined with piroxicam. CONCLUSIONS: Cannabidiol showed promising results as a single agent or in combination with mitoxantrone and vinblastine for treatment of canine urothelial carcinoma cells. Further studies are justified to investigate whether these results are translatable in vivo.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cannabidiol/farmacología , Enfermedades de los Perros/tratamiento farmacológico , Piroxicam/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/farmacología , Anticonvulsivantes/farmacología , Apoptosis , Carboplatino/administración & dosificación , Supervivencia Celular , Enfermedades de los Perros/patología , Perros , Quimioterapia Combinada , Mitoxantrona/administración & dosificación , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/patología
5.
Vet Comp Oncol ; 18(4): 559-569, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32060984

RESUMEN

Osteosarcoma (OSA) is the most common primary bone cancer in children, adolescents and dogs. Current combination surgical and chemotherapeutic treatments have increased survival. However, in recurrent or metastatic disease settings, the prognosis significantly decreases, representing an urgent need for better second-line and novel chemotherapeutics. The current gold standard for combination chemotherapy in OSA often includes a platinum agent, for example, cisplatin or carboplatin. These platinum agents are shuttled within the cell via copper transporters. Recent interest in targeting copper transport has been directed towards antioxidant protein 1 (Atox1) and copper chaperone for superoxide dismutase 1 (CCS), with Atox1 demonstrating the ability to aggregate platinum agents, preventing them from forming DNA adducts. DC_AC50 is a small molecule inhibitor of both Atox1 and CCS. To assess the impact of targeting these pathways on chemotherapy response, two human and two canine OSA cell lines were utilized. After treatment with single agent or combination drugs, cell viability was evaluated and pharmacological synergism calculated using the combination index method. Apoptosis, cell cycle distribution, clonogenic survival and migration were also evaluated. DC_AC50 synergised with carboplatin in combination treatment of human and canine OSA cells to reduce cancer cell viability. DC_AC50-treated cells were significantly less mitotically active, as demonstrated by decreased expression of phospho-histone H3 and cell cycle analysis. DC_AC50 also potentiated carboplatin-induced apoptosis in OSA cells and decreased clonogenic survival. Finally, DC_AC50 reduced the migratory ability of OSA cells. These results justify further investigation into inhibiting intracellular copper chaperones as a means of reducing/preventing acquired chemotherapy resistance.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/veterinaria , Carboplatino/farmacología , Enfermedades de los Perros/tratamiento farmacológico , Osteosarcoma/veterinaria , Animales , Neoplasias Óseas/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cobre , Enfermedades de los Perros/patología , Perros , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada/métodos , Quimioterapia Combinada/veterinaria , Humanos , Chaperonas Moleculares , Osteosarcoma/tratamiento farmacológico , Peroxiredoxina III/efectos de los fármacos
6.
J Vet Intern Med ; 33(2): 800-811, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30556178

RESUMEN

BACKGROUND: Osteosarcoma (OSA) is a common bone tumor of mesenchymal origin in dogs. Chemotherapy delays metastasis, yet most dogs die of this disease within 1 year of diagnosis. The high metabolic demand of cancer cells promotes proton pump upregulation, leading to acidification of the tumor microenvironment and chemoresistance. The potassium-sparing diuretic amiloride is among a class of proton pump inhibitors prescribed for refractory heart failure treatment in dogs. OBJECTIVE: We hypothesized that amiloride treatment improves chemotherapy response by reducing acidification in canine OSA cells. Our objective was to assess the in vitro effects of amiloride on cell viability, apoptosis, and metabolism. METHODS: In vitro study. Assessments of cell viability and apoptosis were performed after single agent or combination treatment, along with calculations of pharmacological synergism using the combination index. Protein signaling during apoptosis was evaluated by Western blotting. Metabolic profiling was performed using a Seahorse bioanalyzer. RESULTS: Amiloride strongly synergized with doxorubicin in combination treatment and exhibited additive or antagonistic effects with carboplatin in canine OSA cells. Combination treatment with doxorubicin significantly upregulated p53-mitochondrial signaling to activate apoptosis and downregulate Akt phosphorylation. Amiloride-treated cells further exhibited metabolic switching with reductions in glycolytic capacity and maximal respiration. CONCLUSION AND CLINICAL IMPORTANCE: Amiloride synergized with doxorubicin to potentiate apoptosis in canine OSA cells. These results justify further investigation into repurposing of amiloride as an oncology drug for the treatment of OSA in dogs.


Asunto(s)
Amilorida/uso terapéutico , Antineoplásicos/uso terapéutico , Carboplatino/uso terapéutico , Doxorrubicina/uso terapéutico , Osteosarcoma/veterinaria , Inhibidores de la Bomba de Protones/uso terapéutico , Amilorida/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/veterinaria , Carboplatino/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Enfermedades de los Perros/tratamiento farmacológico , Perros , Doxorrubicina/administración & dosificación , Quimioterapia Combinada/veterinaria , Osteosarcoma/tratamiento farmacológico , Inhibidores de la Bomba de Protones/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...