Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Int Immunopharmacol ; 112: 109278, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36215870

ABSTRACT

Inflammatory bone destruction has gradually attracted attention worldwide and has been observed in several kinds of pathological bone diseases, such as osteoarthritis, osteomyelitis, rheumatic arthritis, and other infectious clinical trials in the skeletal system. In this regard, excessive osteoclasts and bone resorption activity participate in osteolytic processes. Thus, negatively modulating osteoclast differentiation and bone erosion has been considered an effective therapeutic strategy to limit the poor progression of inflammatory osteolysis. Astragalin (AST) is a bioactive component of traditional Chinese drugs, such as Rosa agrestis, which presents anti-inflammatory and antioxidant effects. However, it is unclear how AST may play an essential role in regulating the dynamic balance of the bone matrix by affecting osteoclastogenesis. This study found that AST could inhibit osteoclastic formation and bone resorption activity in a dose-dependent manner without cytotoxicity. Administration of AST also inhibited the expression of cathepsin K, c-Fos, NFATc1, and TRAP at different stages of mRNA and protein levels during osteoclastogenesis. Reactive oxygen species (ROS) signalling could also be modulated by treatment with AST during RANKL-induced osteoclast differentiation through the Nrf2-HO1 signalling pathway. Additionally, AST could negatively regulate mitogen-activated protein kinase (MAPK) signalling in this process. In vivo, AST significantly reduced lipopolysaccharide (LPS)-induced bone loss in an osteolytic mouse model. AST might be a promising therapeutic candidate for treating osteolytic bone diseases in the future.


Subject(s)
Bone Resorption , Osteolysis , Mice , Animals , Osteolysis/metabolism , Osteogenesis , Reactive Oxygen Species/metabolism , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Cathepsin K/metabolism , NF-E2-Related Factor 2/metabolism , Antioxidants/therapeutic use , RANK Ligand/metabolism , Osteoclasts , Bone Resorption/pathology , Signal Transduction , Anti-Inflammatory Agents/therapeutic use , RNA, Messenger/metabolism , Cell Differentiation
2.
Front Immunol ; 13: 993187, 2022.
Article in English | MEDLINE | ID: mdl-36119068

ABSTRACT

Patients with stage III lung adenocarcinoma (LUAD) have significant survival heterogeneity, meanwhile, CD8+ T cell has a remarkable function in immunotherapy. Therefore, developing novel biomarkers based on CD8+ T cell can help evaluate the prognosis and guide the strategy of immunotherapy for patients with stage III LUAD. Thus, we abstracted twelve datasets from multiple online databases and grouped the stage III LUAD patients into training and validation sets. We then used WGCNA and CIBERSORT, while univariate Cox analysis, LASSO analysis, and multivariate Cox analysis were performed. Subsequently, a novel CD8+ T cell-related classifier including HDFRP3, ARIH1, SMAD2, and UPB1 was developed, which could divide stage III LUAD patients into high- and low-risk groups with distinct survival probability in multiple cohorts (all P < 0.05). Moreover, a robust nomogram including the traditional clinical parameters and risk signature was constructed, and t-ROC, C-index, and calibration curves confirmed its powerful predictive capacity. Besides, we detected the difference in immune cell subpopulations and evaluated the potential benefits of immunotherapy between the two risk subsets. Finally, we verified the correlation between the gene expression and CD8+ T cells included in the model by immunohistochemistry and validated the validity of the model in a real-world cohort. Overall, we constructed a robust CD8+ T cell-related risk model originally which could predict the survival rates in stage III LUAD. What's more, this model suggested that patients in the high-risk group could benefit from immunotherapy, which has significant implications for accurately predicting the effect of immunotherapy and evaluating the prognosis for patients with stage III LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/therapy , CD8-Positive T-Lymphocytes/metabolism , Humans , Immunotherapy , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Prognosis , Ubiquitin-Protein Ligases
3.
Immunotherapy ; 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35259921

ABSTRACT

Aim: To compare the efficacy and safety of first-line chemotherapy (Chemo) plus immune checkpoint inhibitors (ICIs) or bevacizumab (Bev) in advanced non-squamous non-small-cell lung cancer without EGFR mutations or ALK fusions. Methods: A network meta-analysis was conducted to synthesize relative treatment outcomes. Results: Chemo + ICIs is superior to Chemo + Bev in both overall survival (hazard ratio: 0.92; 95% CI: 0.88-0.96) and progression-free survival (hazard ratio: 0.93; 95% CI: 0.90-0.97), with comparable severe adverse events. However, for patients with liver metastasis, Chemo + Bev has a 59.8% probability of providing better overall survival benefit. For specific regimens, pembrolizumab + Chemo showed an absolute advantage over other regimens. Conclusion: First-line Chemo + ICIs is superior to Chemo + Bev in advanced non-squamous non-small-cell lung cancer except for patients with liver metastasis.


Chemotherapy plus immune checkpoint inhibitors and chemotherapy plus bevacizumab were both superior to traditional chemotherapy and were recommended as the first-line treatment for advanced non-squamous non-small-cell lung cancer patients without EGFR mutations or ALK fusions. However, the efficacy and safety of these two treatment models have not been comprehensively discussed head-to-head in clinical trials. Therefore, a network meta-analysis was performed to compare efficacy between these two treatment models, especially according to different clinical characteristics, to guide decision making in clinical practice.

4.
Front Oncol ; 11: 698199, 2021.
Article in English | MEDLINE | ID: mdl-34589422

ABSTRACT

BACKGROUND: Nivolumab + ipilimumab + two cycles chemotherapy (N-I + chemo, intensive immunotherapy but chemo-light) and pembrolizumab + chemotherapy (Pem + chemo) were both recommended as first-line treatment for metastatic non-small cell lung carcinoma (NSCLC) patients. We conducted this indirect comparison to compare the efficacy of and safety between these two treatments for providing reference for decision making. METHODS: Relevant databases were searched for eligible trials. A well-accepted adjusted indirect treatment comparison (ITC) approach was selected to pool efficacy results and safety outcomes. Subgroup analyses were stratified according to PD-L1 expression and clinical characteristics. RESULTS: Four eligible randomized trials (CheckMate9LA, KEYNOTE-021G, KEYNOTE 189, KEYNOTE 407) involving 2017 patients were available to analyze. The ITC results suggested that N-I + chemo is comparable to Pem + chemo in OS (HR 1.03, 95% CI 0.82-1.30) and ORR (RR 0.81, 95% CI 0.62-1.06), but tended to yield inferior PFS (HR 1.28, 95% CI 1.04-1.59) than did Pem + chemo. As for safety profiles, N-I + chemo showed no significant difference relative to Pem + chemo in any grade adverse events: (RR 1.03, 95% CI 0.99-1.10), but demonstrated reduced toxicity in chemo-related adverse events, such as anemia (RR 0.63, 95% CI 0.49-0.81), neutropenia (RR0.51, 95% CI 0.33-0.79), and thrombocytopenia (RR 0.38, 95% CI 0.21-0.69). CONCLUSIONS: N-I + chemo is a promising treatment option for providing comparable OS related to Pem + chemo. However, for never smoker female patients, Pem + chemo is preferable to choose for demonstrating favorable OS benefit than N-I + chemo.

5.
J Exp Clin Cancer Res ; 39(1): 284, 2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33317597

ABSTRACT

The enthusiasm for immune checkpoint inhibitors (ICIs), an efficient tumor treatment model different from traditional treatment, is based on their unprecedented antitumor effect, but the occurrence of immune-related adverse events (irAEs) is an obstacle to the prospect of ICI treatment. IrAEs are a discrete toxicity caused by the nonspecific activation of the immune system and can affect almost all tissues and organs. Currently, research on biomarkers mainly focuses on the gastrointestinal tract, endocrine system, skin and lung. Several potential hypotheses concentrate on the overactivation of the immune system, excessive release of inflammatory cytokines, elevated levels of pre-existing autoantibodies, and presence of common antigens between tumors and normal tissues. This review lists the current biomarkers that might predict irAEs and their possible mechanisms for both nonspecific and organ-specific biomarkers. However, the prediction of irAEs remains a major clinical challenge to screen and identify patients who are susceptible to irAEs and likely to benefit from ICIs.


Subject(s)
Biomarkers/metabolism , Drug-Related Side Effects and Adverse Reactions/diagnosis , Immune Checkpoint Inhibitors/adverse effects , Neoplasms/drug therapy , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/metabolism , Humans , Neoplasms/immunology , Neoplasms/pathology
6.
Pathol Res Pract ; 216(9): 153103, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32825967

ABSTRACT

BACKGROUND: Laryngeal squamous cell carcinoma (LSCC) accounts for about 85%-90% of all cases of laryngeal cancer. So far, the role and molecular mechanism of circular RNA 0,000,218 (circ_0000218)/microRNA (miR)-139-3p in laryngeal cancer are not clear. The present study aimed to investigate the role and regulatory mechanism of circ_0000218/miR-139-3p in laryngeal cancerin vitro and in vivo. METHODS: quantitative real time polymerase chain reaction (qRT-PCR) was used to detect the expression of circ_0000218/miR-139-3p in LSCC cells. Dual luciferase reporter assay and RNA immunoprecipitation (RIP) assay were used to confirm binding sites between miR-139-3p and smad family member 3 (Smad3), and circ_0000218 and miR-139-3p. Cell Counting Kit-8 (CCK-8) and cell apoptosis analysis were used to detect cell viability and apoptosis. Xenograft experiment was performed to show in vivo effect of circ_0000218/miR-139-3p on the growth of LSCC. RESULTS: Circ_0000218 was highly expressed in LSCC cells. miR-139-3p, lower expressed in LSCC cells, was negatively regulated by circ_0000218 in LSCC cells. Besides, the findings suggested that circ_0000218 silencing inhibited the LSCC cell viability and promoted apoptosis by negatively regulating miR-139-3p expression. Furthermore, the data indicated that miR-139-3p inhibited the viability of LSCC cells and promoted apoptosis, and these effects were reversed by Smad3 over-expression. In addition, the in vivo effects of circ_0000218/miR-139-3p on LSCC were consistent with the in vitro study. CONCLUSIONS: circ_0000218 inhibition inhibited the growth of LSCC by targeting miR-139-3p/Smad3 axis. Our present study provided a new target for laryngeal cancer treatment.


Subject(s)
Laryngeal Neoplasms/genetics , MicroRNAs/genetics , RNA, Circular/genetics , Smad3 Protein/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinogenesis/genetics , Carcinogens/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/pathology
7.
Front Genet ; 11: 899, 2020.
Article in English | MEDLINE | ID: mdl-32849853

ABSTRACT

Malignant pleural mesothelioma (MPM), predominantly caused by asbestos exposure, is a highly aggressive cancer with poor prognosis. The staging systems currently used in clinics is inadequate in evaluating the prognosis of MPM. In this study, a five-gene signature was developed and enrolled into a prognostic risk score model by LASSO Cox regression analysis based on two expression profiling datasets (GSE2549 and GSE51024) from Gene Expression Omnibus (GEO). The five-gene signature was further validated using the Cancer Genome Atlas (TCGA) MPM dataset. Univariate and multivariate Cox analyses proved that the five-gene signature was an independent prognostic factor for MPM. The signature remained statistically significant upon stratification by Brigham stage, AJCC stage, gender, tumor size, and lymph node status. Time-dependent receiver operating characteristic (ROC) curve indicated good performance of our model in predicting 1- and 2-years overall survival in MPM patients. The C-index was 0.784 for GSE2549 and 0.753 for the TCGA dataset showing moderate predictive accuracy of our model. Furthermore, Gene Set Enrichment Analysis suggested that the five-gene signature was related to pathways resulting in MPM tumor progression. Together, we have established a five-gene signature significantly associated with prognosis in MPM patients. Hence, the five-genes signature may serve as a potentially useful prognostic tool for MPM patients.

8.
Lung Cancer ; 147: 143-153, 2020 09.
Article in English | MEDLINE | ID: mdl-32717571

ABSTRACT

OBJECTIVE: Progress in neoadjuvant therapy for resectable nonsmall cell lung cancer(NSCLC1)has been stagnant. There have been great achievements in immunotherapy for advanced NSCLC, but the efficacy and safety of neoadjuvant immunotherapy for resectable NSCLC has not been clearly demonstrated. METHODS: Original articles describing the safety and efficacy of neoadjuvant immunotherapy in resectable NSCLC published before January 2020 were retrieved from PubMed, Embase and Cochrane Library. The odds ratio (OR2) and 95 % confidence interval (CI3) were calculated. Heterogeneity and subgroup analysis were performed. RESULTS: A total of 252 patients from seven studies were included. Major pathological response (MPR4) and pathological complete response (pCR5) were used to evaluate the efficacy of neoadjuvant immunotherapy. Compared with neoadjuvant chemotherapy, which exhibited less than 25 % MPR and approximately 2 %-15 % pCR, the values in neoadjuvant immunotherapy were significantly higher (MPR: OR = 0.59; 95 % CI, 0.36-0.98; pCR: OR = 0.16; 95 % CI, 0.09-0.27). Safety was evaluated by the incidence of treatment-related adverse events (TRAE6), surgical resection rate, incidence of surgical complications and surgical delay rate. The pooled OR values of the incidence of TRAE, incidence of surgical complications and surgical delay rate were 0.19, 0.41 and 0.03, respectively, which were significantly better than those for neoadjuvant chemotherapy (95 % CI, 0.04-0.90; 0.22-0.75; 0.01-0.10, respectively). The mean surgical resection rate was 88.70 %, which was similar to the 75 %-90 % rate reported for neoadjuvant chemotherapy (OR = 7.61; 95 % CI, 4.90-11.81). CONCLUSION: Neoadjuvant immunotherapy is effective and safe for resectable NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/therapy , Humans , Immunotherapy , Lung Neoplasms/therapy , Neoadjuvant Therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...