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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
BMC Cancer ; 23(1): 1244, 2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38104105

RESUMO

AIMS: To investigate the predictive value of baseline C-reactive protein (CRP) levels on the efficacy of chemotherapy plus immune checkpoint inhibitors (ICI) in patients with advanced lung squamous cell carcinoma (LSCC). MATERIALS AND METHODS: In this retrospective multicenter study spanning from January 2016 to December 2020, advanced LSCC patients initially treated with chemotherapy or a combination of chemotherapy and ICI were categorized into normal and elevated CRP subgroups. The relationship between CRP levels and treatment outcomes was analyzed using multivariate Cox proportional hazards models and multivariate logistic regression, focusing primarily on the progression-free survival (PFS) endpoint, and secondarily on overall survival (OS) and objective response rate (ORR) endpoints. Survival curves were generated using the Kaplan-Meier method, with the log-rank test used for comparison between groups. RESULTS: Of the 245 patients evaluated, the 105 who received a combination of chemotherapy and ICI with elevated baseline CRP levels exhibited a significant reduction in PFS (median 6.5 months vs. 11.8 months, HR, 1.78; 95% CI: 1.12-2.81; p = 0.013) compared to those with normal CRP levels. Elevated CRP was identified as an independent risk factor for poor PFS through multivariate-adjusted analysis. However, among the 140 patients receiving chemotherapy alone, baseline CRP levels did not significantly influence PFS. Furthermore, within the combination therapy group, there was a notable decrease in the ORR (51% vs. 71%, p = 0.035), coupled with a significantly shorter OS (median 20.9 months vs. 31.5 months, HR, 2.24; 95% CI: 1.13-4.44; p = 0.033). CONCLUSION: In patients with advanced LSCC, elevated baseline CRP levels were identified as an independent predictive factor for the efficacy of combination therapy with chemotherapy and ICI, but not in chemotherapy alone. This suggests that CRP may be a valuable biomarker for guiding treatment strategies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Humanos , Proteína C-Reativa , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Estudos Retrospectivos , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Pulmão
2.
Clin Cancer Res ; 25(5): 1546-1556, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30389658

RESUMO

PURPOSE: Examining the role of developmental signaling pathways in "driver gene-negative" lung adenocarcinoma (patients with lung adenocarcinoma negative for EGFR, KRAS, BRAF, HER2, MET, ALK, RET, and ROS1 were identified as "driver gene-negative") may shed light on the clinical research and treatment for this lung adenocarcinoma subgroup. We aimed to investigate whether developmental signaling pathways activation can stratify the risk of "driver gene-negative" lung adenocarcinoma. EXPERIMENTAL DESIGN: In the discovery phase, we profiled the mRNA expression of each candidate gene using genome-wide microarrays in 52 paired lung adenocarcinoma and adjacent normal tissues. In the training phase, tissue microarrays and LASSO Cox regression analysis were applied to further screen candidate molecules in 189 patients, and we developed a predictive signature. In the validation phase, one internal cohort and two external cohorts were used to validate our novel prognostic signature. RESULTS: Kyoto Encyclopedia of Genes and Genomes pathway analysis based on whole-genome microarrays indicated that the Wnt/ß-catenin pathway was activated in "driver gene-negative" lung adenocarcinoma. Furthermore, the Wnt/ß-catenin pathway-based gene expression profiles revealed 39 transcripts differentially expressed. Finally, a Wnt/ß-catenin pathway-based CSDW signature comprising 4 genes (CTNNB1 or ß-catenin, SOX9, DVL3, and Wnt2b) was developed to classify patients into high-risk and low-risk groups in the training cohort. Patients with high-risk scores in the training cohort had shorter overall survival [HR, 10.42; 6.46-16.79; P < 0.001) than patients with low-risk scores. CONCLUSIONS: The CSDW signature is a reliable prognostic tool and may represent genes that are potential drug targets for "driver gene-negative" lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/etiologia , Adenocarcinoma de Pulmão/mortalidade , Proteínas Oncogênicas/genética , Transcriptoma , Adenocarcinoma de Pulmão/diagnóstico , Adenocarcinoma de Pulmão/metabolismo , Adulto , Idoso , Biomarcadores Tumorais , Biópsia , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Via de Sinalização Wnt
3.
Gene ; 576(1 Pt 1): 22-7, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26432005

RESUMO

The metabolic program of cancer cells is significant different from the normal cells, which makes it possible to develop novel strategies targeting cancer cells. Mevalonate pathway and its rate-limiting enzyme HMG-CoA reductase (HMGCR) have shown important roles in the progression of several cancer types. However, their roles in glioblastoma cells remain unknown. In this study, up-regulation of HMGCR in the clinical glioblastoma samples was observed. Forced expression of HMGCR promoted the growth and migration of U251 and U373 cells, while knocking down the expression of HMGCR inhibited the growth, migration and metastasis of glioblastoma cells. Molecular mechanism studies revealed that HMGCR positively regulated the expression of TAZ, an important mediator of Hippo pathway, and the downstream target gene connective tissue growth factor (CTGF), suggesting HMGCR might activate Hippo pathway in glioblastoma cells. Taken together, our study demonstrated the oncogenic roles of HMGCR in glioblastoma cells and HMGCR might be a promising therapeutic target.


Assuntos
Movimento Celular , Proliferação de Células , Glioblastoma/enzimologia , Hidroximetilglutaril-CoA Redutases/metabolismo , Proteínas de Neoplasias/metabolismo , Aciltransferases , Linhagem Celular Tumoral , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Glioblastoma/genética , Glioblastoma/patologia , Células HEK293 , Humanos , Hidroximetilglutaril-CoA Redutases/genética , Metástase Neoplásica , Proteínas de Neoplasias/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Am J Transl Res ; 7(10): 2026-35, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26692946

RESUMO

Cigarette smoking represents for the highest risk-factor for non-small cell lung cancer (NSCLC), and a growing body of evidence suggested that smoking was associated with a high recurrence and poor therapeutic response of NSCLC as well. On the other hand, epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs), such as gefitinib, has been proved to be an efficient and safe strategy for treating NSCLC. Although accumulating clinical data suggested that smoking history might influence the therapeutic effects of EGFR-TKIs even in NSCLC patients harboring sensitive EGFR mutation, the exact effects of cigarette smoking on the efficacy of EGFR-TKIs treatment in NSCLC patients remain exclusive. In this study, we firstly identified the adverse effect of smoking exposure on the efficacy of EGFR-TKIs treatment against lung adenocarcinoma in mutation-positive patients by retrospective analysis of clinical data. The hypo-responsiveness of smoking patients on the therapy was accompanied with persistent activation of EGFR-downstream signal molecules ERK1/2 and AKT, which could not be inhibited by gefitinib and thus lead to the failure of EGFR-TKIs treatment. Based on our in vitro data, it was also found that long-term cigarette smoking extract (CSE) exposure induced epithelial-mesenchymal transition (EMT), which might also contribute to acquired resistance to EGFR-TKIs. Taken together, our findings suggested that cigarette smoking negatively regulated the clinical outcome of EGFR-TKIs therapy in lung adenocarcinoma patients, which was correlated with the activation of EGFR signaling and the induction of EMT.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA