Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Base de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancers (Basel) ; 16(14)2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39061136

RESUMEN

CAR-T cell-based therapies have demonstrated remarkable efficacy in treating malignant cancers, especially liquid tumors, and are increasingly being evaluated in clinical trials for solid tumors. With the FDA's initiative to advance alternative methods for drug discovery and development, full human ex vivo assays are increasingly essential for precision CAR-T development. However, prevailing ex vivo CAR-T cell-mediated cytotoxicity assays are limited by their use of radioactive materials, lack of real-time measurement, low throughput, and inability to automate, among others. To address these limitations, we optimized the assay using multimodality imaging methods, including bioluminescence, impedance tracking, phase contrast, and fluorescence, to track CAR-T cells co-cultured with CD19, CD20, and HER2 luciferase reporter cancer cells in real-time. Additionally, we varied the ratio of CAR-T cells to cancer cells to determine optimal cytotoxicity readouts. Our findings demonstrated that the CAR-T cell group effectively attacked cancer cells, and the optimized assay provided superior temporal and spatial precision measurements of ex vivo CAR-T killing of cancer cells, confirming the reliability, consistency, and high throughput of the optimized assay.

2.
Cancers (Basel) ; 16(11)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38893085

RESUMEN

Recent studies highlight the integral role of the interferon gamma receptor (IFNγR) pathway in T cell-mediated cytotoxicity against solid but not liquid tumors. IFNγ not only directly facilitates tumor cell death by T cells but also indirectly promotes cytotoxicity via myeloid phagocytosis in the tumor microenvironment. Meanwhile, full human ex vivo immune checkpoint drug screening remains challenging. We hypothesized that an engineered gamma interferon activation site response element luciferase reporter (GAS-Luc2) can be utilized for immune checkpoint drug screening in diverse ex vivo T cell-solid tumor cell co-culture systems. We comprehensively profiled cell surface proteins in ATCC's extensive collection of human tumor and immune cell lines, identifying those with endogenously high expression of established and novel immune checkpoint molecules and binding ligands. We then engineered three GAS-Luc2 reporter tumor cell lines expressing immune checkpoints PD-L1, CD155, or B7-H3/CD276. Luciferase expression was suppressed upon relevant immune checkpoint-ligand engagement. In the presence of an immune checkpoint inhibitor, T cells released IFNγ, activating the JAK-STAT pathway in GAS-Luc2 cells, and generating a quantifiable bioluminescent signal for inhibitor evaluation. These reporter lines also detected paracrine IFNγ signaling for immune checkpoint-targeted ADCC drug screening. Further development into an artificial antigen-presenting cell line (aAPC) significantly enhanced T cell signaling for superior performance in these ex vivo immune checkpoint drug screening platforms.

3.
Cancers (Basel) ; 14(21)2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-36358868

RESUMEN

BRAF V600E mutation drives uncontrolled cell growth in most melanomas. While BRAF V600E tumors are initially responsive to BRAF inhibitors, prolonged treatment results in inhibitor resistance and tumor regrowth. Clinical data have linked the NRAS Q61K, KRAS G13D and MEK1 Q56P mutations to the BRAF inhibitor resistance. However, development of novel therapeutics is hindered by the lack of relevant isogeneic cell models. We employed CRISPR/Cas9 genome engineering to introduce NRAS Q61K, KRAS G13D and MEK1 Q56P mutations into the A375 melanoma cell line with endogenously high expression of BRAF V600E. The resulting isogenic cell lines are resistant to BRAF inhibitors. The A375 MEK1 Q56P isogenic cells are additionally resistant to MEK inhibitors as single agent, but interestingly, these cells become sensitive to MEK/BRAF inhibitor combo. Our results suggest that resistance in the NRAS and MEK isogenic lines is driven by constitutive MEK/ERK signaling, while the resistance in the KRAS isogenic line is driven by EGFR overexpression. Interestingly, the KRAS G13D isogenic line displays elevated PD-L1 expression suggesting the KRAS G13D mutation could be a potential indication for immunotherapy. Overall, these three novel isogenic cell models with endogenous level RAS and MEK1 point mutations provide direct bio-functional evidence demonstrating that acquiring a drug-resistant gene drives tumor cell survival and may simultaneously introduce new indications for combo therapy or immunotherapy in the clinic.

4.
Cancer Biol Ther ; 5(10): 1361-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16969069

RESUMEN

Downregulation of the EGF receptor is the net result of receptor degradation and recycling. Cbl functions by specifically targeting activated ErbB receptors for ubiquitination, facilitating ligand-induced desensitization of EGFR. The interaction between EGFR and c-Cbl has been shown to depend upon receptor phosphorylation at tyrosine residue 1045, the major docking site for c-Cbl. To better understand the biological consequences of EGFR mutants in human cancers, we compared wild-type EGFR and EGFRvIII internalization, as well as gefitinib sensitive and resistant EGFR kinase mutations found in non-small cell lung carcinoma. We observed that Cbl failed to associate with EGFRvIII as well as an inability of the receptor to undergo ubiquitination and degradation. The most intriguing observation is that EGFRvIII tyrosine 1045 residue is either un-phosphorylated or hypophosphorylated. This is in contrast to other tyrosine residues in EGFRvIII, such as Y1173, which exhibit levels of phosphorylation comparable to those of wild-type EGFR. These results suggest that hypophosphorylation of tyrosine residue 1045 is likely to be the cause for EGFRvIII escape from c-Cbl-induced ubiquitination and degradation, enhancing EGFRvIII's ability to increase proliferation in breast cancer cells. Interestingly, inefficient degradation was only observed in the gefitinib resistant EGFR kinase mutant, despite the fact that this mutant receptor is capable of recruiting c-Cbl and undergoes ubiquitination. The gefitinib sensitive EGFR kinase mutant exhibits similar ubiquitination and degradation patterns as the wild-type EGFR. Collectively, different EGFR mutations exert various negative mechanisms that have the potential to modify receptor internalization and degradation, and may play a critical role in resistance to tyrosine kinase inhibitory treatments.


Asunto(s)
Receptores ErbB/fisiología , Regulación Neoplásica de la Expresión Génica , Fosfotirosina/metabolismo , Secuencia de Aminoácidos , Neoplasias de la Mama , Carcinoma de Pulmón de Células no Pequeñas , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Neoplasias Pulmonares , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA