Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Thorac Oncol ; 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38553005

RESUMEN

INTRODUCTION: EGFR-mutated NSCLC is characterized by an immunosuppressive microenvironment that confers limited clinical effectiveness to anti-PD-1 or PD-L1 antibodies. Despite the discouraging outcomes of immunotherapy, novel immune checkpoints are constantly emerging, among which the specific vulnerability for therapeutic intervention in the context of EGFR-mutated NSCLC remains unresolved. METHODS: Data sets of patient- and cell line-levels were used for screening and mutual validation of association between EGFR mutation and a panel of immune checkpoint-related genes. Regulatory mechanism was elucidated through in vitro manipulation of EGFR signaling pathway and evaluated by immunoblot analysis, quantitative polymerase chain reaction, flow cytometry, immunofluorescence staining, and chromatin immunoprecipitation. In vivo investigation of different therapeutic strategies were conducted using both immunocompetent and immunodeficient mouse models. RESULTS: Among all screened immune checkpoints, CD47 emerged as the candidate most relevant to EGFR activation. Mechanistically, EGFR mutation constitutively activated downstream ERK and AKT pathways to respectively up-regulate the transcriptional factors c-Myc and NF-κB, both of which structurally bound to the promotor region of CD47 and actively transcribed this "don't eat me" signal. Impaired macrophage phagocytosis was observed on introduction of EGFR-sensitizing mutations in NSCLC cell line models, whereas CD47 blockade restored the phagocytic capacity and augmented tumor cell killing in both in vitro and in vivo models. Remarkably, the combination of anti-CD47 antibody with EGFR tyrosine kinase inhibitor revealed an additive antitumor activity compared with monotherapy of either antitumor agent in both immunocompetent and adaptive immunity-deficient mouse models. CONCLUSIONS: EGFR-sensitizing mutation facilitates NSCLC's escape from innate immune attack through up-regulating CD47. Combination therapy incorporating CD47 blockade holds substantial promise for clinical translation in developing more effective therapeutic approaches against EGFR-mutant NSCLC.

2.
Acta Biomater ; 161: 184-200, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36893957

RESUMEN

Non-small cell lung cancer (NSCLC) remains the most frequently diagnosed lung cancer and the leading cause of cancer-related mortality worldwide. PD-1/PD-L1 axis inhibitors have changed the treatment paradigm for various cancer types, including NSCLC. However, success of these inhibitors in lung cancer clinic is severely limited by their inability to inhibit the PD-1/PD-L1 signaling axis due to heavy glycosylation and heterogeneity expression of PD-L1 in NSCLC tumor tissue. Taking advantage of the facts that tumor cell derived nanovesicles could efficiently accumulate in the homotypic tumor sites due to their innate targeting abilities and that specific and high affinity existed between PD-1 and PD-L1, we developed NSCLC targeting biomimetic nanovesicles (NV) cargos from genetically engineered NSCLC cell lines that overexpressed PD-1 (P-NV). We showed that P-NVs efficiently bound NSCLC cells in vitro and targeted tumor nodules in vivo. We further loaded P-NVs with 2-deoxy-D-glucose (2-DG) and doxorubicin (DOX), and found that these drugs co-loaded P-NVs efficiently shrank lung cancers in mouse models for both allograft and autochthonous tumor. Mechanistically, drug-loaded P-NVs efficiently caused cytotoxicity to tumor cells and simultaneously activated anti-tumor immunity function of tumor-infiltrating T cells. Our data therefore strongly argue that 2-DG and DOX co-loaded, PD-1-displaying nanovesicles is a highly promising therapy for treatment of NSCLC in clinic. STATEMENT OF SIGNIFICANCE: Lung cancer cells overexpressing PD-1 are developed for preparing nanoparticles (P-NV). PD-1s displayed on NVs enhance their homologous targeting abilities to tumor cells expressing PD-L1s. Chemotherapeutics such as DOX and 2-DG, are packaged in such nanovesicles (PDG-NV). These nanovesicles efficiently delivered chemotherapeutics to tumor nodules specifically. The synergy between DOX and 2-DG is observed in inhibiting lung cancer cells in vitro and in vivo. Importantly, 2-DG causes deglycosylation and downregulation of PD-L1 on tumor cells while PD-1 displayed on nanovesicles' membrane blocks PD-L1 on tumor cells. 2-DG loaded nanoparticles thus activate anti-tumor activities of T cells in the tumor microenvironment. Our work thus highlights the promising antitumor activity of PDG-NVs, which warrants further clinical evaluation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Ratones , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/patología , Antígeno B7-H1 , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/uso terapéutico , Inmunoterapia , Doxorrubicina/uso terapéutico , Línea Celular Tumoral , Microambiente Tumoral
3.
EMBO Mol Med ; 14(1): e14502, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34898004

RESUMEN

Impressive clinical benefit is seen in clinic with PD-1 inhibitors on portion of cancer patients. Yet, there remains an urgent need to develop effective synergizers to expand their clinical application. Tumor-associated macrophage (TAM), a type of M2-polarized macrophage, eliminates or suppresses T-cell-mediated anti-tumor responses. Transforming TAMs into M1 macrophages is an attractive strategy of anti-tumor therapy. Here, we conducted a high-throughput screening and found that Carfilzomib potently drove M2 macrophages to express M1 cytokines, phagocytose tumor cells, and present antigens to T cells. Mechanistically, Carfilzomib elicited unfolded protein response (UPR), activated IRE1α to recruit TRAF2, and activated NF-κB to transcribe genes encoding M1 markers in M2 macrophages. In vivo, Carfilzomib effectively rewired tumor microenvironment through reprogramming TAMs into M1-like macrophages and shrank autochthonous lung cancers in transgenic mouse model. More importantly, Carfilzomib synergized with PD-1 antibody to almost completely regress autochthonous lung cancers. Given the safety profiles of Carfilzomib in clinic, our work suggested a potentially immediate application of combinational treatment with Carfilzomib and PD-1 inhibitors for patients with solid tumors.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares , Oligopéptidos/farmacología , Microambiente Tumoral , Macrófagos Asociados a Tumores/inmunología , Animales , Reprogramación Celular , Endorribonucleasas , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas
4.
Theranostics ; 11(13): 6592-6606, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995678

RESUMEN

Purpose: Clinical success of cancer therapy is severely limited by drug resistance, attributed in large part to the loss of function of tumor suppressor genes (TSGs). Developing effective strategies to treat those tumors is challenging, but urgently needed in clinic. Experimental Design: MYOCD is a clinically relevant TSG in lung cancer patients. Our in vitro and in vivo data confirm its tumor suppressive function. Further analysis reveals that MYOCD potently inhibits stemness of lung cancer stem cells. Mechanistically, MYOCD localizes to TGFBR2 promoter region and thereby recruits PRMT5/MEP50 complex to epigenetically silence its transcription. Conclusions: NSCLC cells deficient of MYOCD are particularly sensitive to TGFBR kinase inhibitor (TGFBRi). TGFBRi and stemness inhibitor synergize with existing drugs to treat MYOCD deficient lung cancers. Our current work shows that loss of function of MYOCD creates Achilles' heels in lung cancer cells, which might be exploited in clinic.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Nucleares/deficiencia , Receptor Tipo II de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Transactivadores/deficiencia , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación hacia Abajo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Código de Histonas , Humanos , Neoplasias Pulmonares/genética , Metilación , Ratones Transgénicos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/patología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Transducción de Señal , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/fisiología , Carga Tumoral
5.
Angew Chem Int Ed Engl ; 59(31): 12868-12875, 2020 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-32289203

RESUMEN

Herein, we report a theoretical and experimental study of the water-gas shift (WGS) reaction on Ir1 /FeOx single-atom catalysts. Water dissociates to OH* on the Ir1 single atom and H* on the first-neighbour O atom bonded with a Fe site. The adsorbed CO on Ir1 reacts with another adjacent O atom to produce CO2 , yielding an oxygen vacancy (Ovac ). Then, the formation of H2 becomes feasible due to migration of H from adsorbed OH* toward Ir1 and its subsequent reaction with another H*. The interaction of Ir1 and the second-neighbouring Fe species demonstrates a new WGS pathway featured by electron transfer at the active site from Fe3+ -O⋅⋅⋅Ir2+ -Ovac to Fe2+ -Ovac ⋅⋅⋅Ir3+ -O with the involvement of Ovac . The redox mechanism for WGS reaction through a dual metal active site (DMAS) is different from the conventional associative mechanism with the formation of formate or carboxyl intermediates. The proposed new reaction mechanism is corroborated by the experimental results with Ir1 /FeOx for sequential production of CO2 and H2 .

6.
Molecules ; 25(7)2020 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-32268564

RESUMEN

Aberrant activation of the WNT/ß-catenin signaling pathway is implicated in various types of cancers. Inhibitors targeting the Wnt signaling pathway are intensively studied in the current cancer research field, the outcomes of which remain to be determined. In this study, we have attempted to discover novel potent WNT/ß-catenin pathway inhibitors through tankyrase 1/2 structure-based virtual screening. After screening more than 13.4 million compounds through molecular docking, we experimentally verified one compound, LZZ-02, as the most potent inhibitor out of 11 structurally representative top hits. LiCl-induced HEK293 cells containing TOPFlash reporter showed that LZZ-02 inhibited the transcriptional activity of ß-catenin with an IC50 of 10 ± 1.2 µM. Mechanistically, LZZ-02 degrades the expression of ß-catenin by stabilizing axin 2, thereby diminishing downstream proteins levels, including c-Myc and cyclin D1. LZZ-02 also inhibits the growth of colonic carcinoma cell harboring constitutively active ß-catenin. More importantly, LZZ-02 effectively shrinks tumor xenograft derived from colonic cell lines. Our study successfully identified a novel tankyrase 1/2 inhibitor and shed light on a novel strategy for developing inhibitors targeting the WNT/ß-catenin signaling axis.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Tanquirasas/antagonistas & inhibidores , Vía de Señalización Wnt/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas c-myb/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Cycle ; 18(23): 3300-3312, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31594449

RESUMEN

Lung cancer is the leading cause of cancer-related deaths worldwide. However, tumor suppressor genes remain to be systemically determined for lung cancer. Here we report interferon regulatory factor 8 (IRF8), a member of the IRF family of transcription factors, as a potent lung tumor suppressor gene. Expression of IRF8 is frequently diminished in lung tumoral tissues and is associated with prognosis of non-small cell lung cancer (NSCLC) patients. Ectopic expression of IRF8 suppresses the NSCLC cells proliferation in vitro and tumorigenic potential in vivo. More importantly, forced expression of IRF8 through infection of recombinant virus inhibits lung tumorigenesis in genetically engineered mouse model (GEMM). Mechanistically, IRF8 inhibits AKT signaling and promotes accumulation of P27 protein, which results in senescence of lung cancer cells. Ectopic expression of IRF8 in tumor cells leads to regression of lung cancer tumor nodules in a xenograft tumor model. Our data, therefore, solidly shows IRF8 to be a lung cancer suppressor gene and may denote an opportunity for therapeutic intervention of NSCLC.


Asunto(s)
Carcinogénesis/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Senescencia Celular/genética , Factores Reguladores del Interferón/genética , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Humanos , Ratones , Pronóstico , Transducción de Señal/genética , Proteínas Supresoras de Tumor/genética
8.
J Am Chem Soc ; 135(41): 15314-7, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24090210

RESUMEN

High specific activity and cost effectiveness of single-atom catalysts hold practical value for water gas shift (WGS) reaction toward hydrogen energy. We reported the preparation and characterization of Ir single atoms supported on FeO(x) (Ir1/FeO(x)) catalysts, the activity of which is 1 order of magnitude higher than its cluster or nanoparticle counterparts and is even higher than those of the most active Au- or Pt-based catalysts. Extensive studies reveal that the single atoms accounted for ∼70% of the total activity of catalysts containing single atoms, subnano clusters, and nanoparticles, thus serving as the most important active sites. The Ir single atoms seem to greatly enhance the reducibility of the FeO(x) support and generation of oxygen vacancies, leading to the excellent performance of the Ir1/FeO(x) single-atom catalyst. The results have broad implications on designing supported metal catalysts with better performance and lower cost.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA