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











Base de dados
Intervalo de ano de publicação
1.
Front Med (Lausanne) ; 10: 1153537, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37138755

RESUMO

Uterine fibroids (UFs) are the most common benign gynecologic tumors in reproductive-aged women. The typical diagnostic strategies of UFs are transvaginal ultrasonography and pathological feature, while molecular biomarkers are considered conventional options in the assessment of the origin and development of UFs in recent years. Here, we extracted the differential expression genes (DEGs) and differential DNA methylation genes (DMGs) of UFs from the Gene Expression Omnibus (GEO) database, GSE64763, GSE120854, GSE45188, and GSE45187. 167 DEGs with aberrant DNA methylation were identified, and further Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed by the relevant R package. We next discerned 2 hub genes (FOS, and TNFSF10) with autophagy involvement by overlapping 167 DEGs and 232 autophagic regulators from Human Autophagy Database. FOS was identified as the most crucial gene through the Protein-Protein Interactions (PPI) network with the correlation of the immune scores. Moreover, the down-regulated expression of FOS in UFs tissue at both mRNA and protein levels was validated by RT-qPCR and immunohistochemistry respectively. The area under the ROC curve (AUC) of FOS was 0.856, with a sensitivity of 86.2% and a specificity of 73.9%. Overall, we explored the possible biomarker of UFs undergoing DNA-methylated autophagy and provided clinicians with a comprehensive assessment of UFs.

2.
Radiother Oncol ; 184: 109681, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37105304

RESUMO

BACKGROUND: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have a significant therapeutic effect in the treatment of advanced non-small-cell lung cancer (NSCLC) with EGFR mutations. However, the acquired resistance greatly limits the survival benefit of EGFR-TKIs for EGFR-mutant NSCLC patients. We aimed to assess the efficacy and safety of stereotactic body radiotherapy (SBRT) plus EGFR-TKIs in these patients. METHODS: In this prospective, randomized, controlled, phase 2 study, participants were recruited from 4 different hospitals in Wuhan, China. Eligible patients were histologically confirmed to have NSCLC with an EGFR-sensitive mutation (19DEL or 21L858R) and diagnosed at stage IV. Patients who had received first-line EGFR-TKIs treatment including gefitinib, erlotinib, and icotinib and achieved stable disease or partial response were enrolled after three months. Eligible participants were randomly assigned (1:1) to receive SBRT plus EGFR-TKIs or EGFR-TKIs treatment alone. In the combination-group, different tumor sites were irradiated at doses ranging from 30-50 Gy in five fractions. Considering the short duration of SBRT, the TKIs were continued during the radiotherapy. The primary endpoint was progression-free survival (PFS), and the secondary endpoints were overall survival (OS) and safety. This study was registered at ClinicalTrials.gov, with the registration number of NCT03595644. RESULTS: Between May 4, 2018 and Dec 20, 2019, 74 patients were screened, of whom 62 patients were enrolled and randomized. The study was closed early with 62/72 patients due to slow accrual. The enrolled patients were randomly assigned to receive SBRT plus EGFR-TKI(n = 31) or EGFR-TKI alone (n = 31). One patient who was randomized to the SBRT plus EGFR-TKI group refused to receive SBRT during the treatment, and, 61 patients were included the modified intention-to-treat (mITT) analysis, with 30 in the SBRT plus EGFR-TKI and 31 in the EGFR-TKI group. As of the clinical cutoff date (Feb 14, 2022), the median follow-up was 29.4 months (IQR 6.9-38.9). The median PFS of the EGFR-TKI group and SBRT combination group was 9.0 vs 17.6 months (hazard ratio [HR] = 0.52, 95% confidence interval [95%CI], 0.31-0.89, P = 0.016). Meanwhile, the median OS was 23.2 vs 33.6 months (HR [95%CI], 0.53(0.30-0.95); P = 0.026). There was no grade 3 or greater toxicity observed in either group, the grade 2 adverse events were 50% in the EGFR-TKIs + SBRT group while the percentage was 45.2% in the EGFR-TKIs group. CONCLUSIONS: The addition of SBRT significantly delayed the onset of acquired resistance to EGFR-TKIs and prolonged the PFS and OS of patients. Radiotherapy of the primary lesion alone might be superior to metastatic sites. Further confirmatory studies are needed to confirm our findings.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Radiocirurgia , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Radiocirurgia/efeitos adversos , Neoplasias Pulmonares/tratamento farmacológico , Estudos Prospectivos , Inibidores de Proteínas Quinases/efeitos adversos , Receptores ErbB/genética , Mutação
3.
Front Oncol ; 13: 1134149, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37064084

RESUMO

Introduction: Retinoic acid-induced 2 (RAI2) was initially related to cell differentiation and induced by retinoic acid. RAI2 has been identified as an emerging tumor suppressor in breast cancer and colorectal cancer. Methods: In this study, we performed systematic analyses of RAI2 in breast cancer. Meta-analysis and Kaplan-Meier survival curves were applied to identify the survival prediction potential of RAI2. Moreover, the association between RAI2 expression and the abundance of six tumor-infiltrating immune cells was investigated by TIMER, including B cells, CD8+ T cells, CD4+ T cells, B cells, dendritic cells, neutrophils, and macrophages. The expression profiles of high and low RAI2 mRNA levels in GSE7390 were compared to identify differentially expressed genes (DEGs) and the biological function of these DEGs was analyzed by R software, which was further proved in GSE7390. Results: Our results showed that the normal tissues had more RAI2 expression than breast cancer tissues. Patients with high RAI2 expression were related to a favorable prognosis and more immune infiltrates. A total of 209 DEGs and 182 DEGs were identified between the expression profiles of high and low RAI2 mRNA levels in the GSE7390 and GSE21653 databases, respectively. Furthermore, Gene Ontology (GO) enrichment indicated that these DEGs from two datasets were both mainly distributed in "biological processes" (BP), including "organelle fission" and "nuclear division". Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis demonstrated that these DEGs from two datasets were both significantly enriched in the "cell cycle". Common hub genes between the DEGs in GSE7390 and GSE21653 were negatively associated with RAI2 expression, including CCNA2, MAD2L1, MELK, CDC20, and CCNB2. Discussions: These results above suggested that RAI2 might play a pivotal role in preventing the initiation and progression of breast cancer. The present study may contribute to understanding the molecular mechanisms of RAI2 and enriching biomarkers to predict patient prognosis in breast cancer.

4.
Biomed Pharmacother ; 156: 113959, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36411663

RESUMO

Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) bring significant benefits to non-small cell lung cancer patients with EGFR mutations, which represent a breakthrough in lung cancer therapy. However, patients will ultimately develop the acquired resistance to the first- or second-generation EGFR-TKIs after a period of treatment, and EGFR T790M mutation is the most common resistant mechanism. The third-generation EGFR-TKIs target T790M mutation and show potent anti-tumor efficacy, especially in central neural system response. Unfortunately, patients inevitably get resistant to the third-generation EGFR-TKIs due to various mechanisms, which can be mainly divided into EGFR-dependent and -independent ones. EGFR-dependent mechanism refers to manifold EGFR mutations while EGFR-independent mechanisms include bypass signal activation, histologic transformation and so on. To precisely address this issue and improve clinical outcomes, various other therapies (e.g. chemotherapy, radiotherapy, etc.) in combination with the third-generation EGFR-TKIs are designed. However, the current results of combination therapies are insufficient and ambiguous, which remain further exploration. Herein, we provide an updated landscape of the third-generation EGFR-TKIs and elaborate on the complex resistant mechanisms. Notably, we summarize the combination therapies with third-generation EGFR-TKIs and discuss their limitations and future perspective, aiming at providing insights to clinicians from bench to bedside.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Receptores ErbB/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Mutação , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
5.
Oncogene ; 41(6): 782-796, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34853466

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the most lethal gastrointestinal malignancies with high mortality. Recurrence develops within only a few years after curative resection and perioperative adjuvant therapy in 30-50% of these patients. Therefore, it is essential to identify postoperative recurrence biomarkers to facilitate selecting the following surveillance and therapeutic strategies. The general transcription factor IIE subunit beta (GTF2E2) is crucial for physiological and pathological functions, but its roles in the aggression and recurrence of ESCC remain ambiguous. In this study, we found that GTF2E2 was highly expressed in ESCC samples, and elevated GTF2E2 expression predicted early recurrence after surgery for ESCC patients. High expression of GTF2E2 associated with more aggressive clinic features and poor prognosis. GTF2E2 promoted the proliferation and mobility of ESCC cells in vitro and in vivo. We further revealed that miR-139-5p repressed GTF2E2 expression by downregulating its mRNA through binding with Argonaute 2 (Ago2). Rescue assays suggested that miR-139-5p affected GTF2E2-mediated ESCC progression. Moreover, GTF2E2 positively interacted with FUS promoter and regulated FUS expression, and the phenotype changes caused by GTF2E2 manipulation were recovered by rescuing FUS expression in ESCC cells. Additionally, we demonstrated that GTF2E2 promotes ESCC cells progression via activation of the AKT/ERK/mTOR pathway. In conclusion, GTF2E2 may serve as a novel biomarker for recurrence after surgery and a potential therapeutic target for ESCC patients, and it promotes ESCC progression via miR-139-5p/GTF2E2/FUS axis.


Assuntos
Carcinoma de Células Escamosas do Esôfago
6.
Curr Opin Oncol ; 33(1): 55-58, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33165003

RESUMO

PURPOSE OF REVIEW: TKI therapy has shown excellent efficacy and favorable tolerability in patients with mutation-positive nonsmall cell lung cancer. However, there is no clear consensus on the role of TKI as induction therapy. In this article, we reviewed recently published studies to analyze the benefits of tyrosine kinase inhibitors, in particular, EGFR TKIs and ALK TKIs, as inducible treatments for NSCLC. RECENT FINDINGS: Several clinical trials have recently presented their latest data, giving analysis of patient's survival benefits and adverse events. Initial results have demonstrated promising efficacy and safety data. Some clinical case reports and retrospective analysis demonstrated that EGFR/ALK TKIs can significantly improve PFS and the rate of radical surgery. However, there was no statistically significant difference in overall survival time of almost all clinical trials. SUMMARY: TKIs are increasingly accepted by clinicians as induction therapy in NSCLC. Many studies have demonstrated that neoadjuvant therapy increases the likelihood of surgery and is associated with good resection rates, as evidenced by high prospective downstaging rates in patients with locally advanced NSCLC. However, the risk of recurrence remains high with no evidence of overall survival benefits being reported. Now that more clinical trials are being conducted and more data will be available for analysis, a clearer and more comprehensive view of what role TKIs play in induction therapy will emerge.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Ensaios Clínicos Fase II como Assunto , Receptores ErbB/antagonistas & inibidores , Humanos , Quimioterapia de Indução , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/cirurgia , Terapia Neoadjuvante , Ensaios Clínicos Controlados Aleatórios como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA