Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biomolecules ; 14(7)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062533

ABSTRACT

In Asian populations with non-small-cell lung cancer (NSCLC), EGFR mutations are highly prevalent, occurring in roughly half of these patients. Studies have revealed that individuals with EGFR mutation typically fare worse with immunotherapy. In patients who received EGFR tyrosine kinase inhibitor (TKI) treatment followed by anti-PD-1 therapy, poor results were observed. The underlying mechanism remains unclear. We used high-resolution flow cytometry and ELISA to detect the circulating level of small extracellular vesicle (sEV) PD-L1 in NSCLC individuals with EGFR mutations before and after receiving TKIs. The secretion amount of sEV PD-L1 of lung cancer cell lines with EGFR mutations under TKI treatment or not were detected using high-resolution flow cytometry and Western blotting. The results revealed that patients harboring EGFR mutations exhibit increased levels of sEV PD-L1 in circulation, which inversely correlated with the presence of CD8+ T cells in tumor tissues. Furthermore, tumor cells carrying EGFR mutations secrete a higher quantity of PD-L1-positive sEVs. TKI treatment appeared to amplify the levels of PD-L1-positive sEVs in the bloodstream. Mutation-induced and TKI-induced sEVs substantially impaired the functionality of CD8+ T cells. Importantly, our findings indicated that EGFR mutations and TKI therapies promote secretion of PD-L1-positive sEVs via distinct molecular mechanisms, namely the HRS and ALIX pathways, respectively. In conclusion, the increased secretion of PD-L1-positive sEVs, prompted by genetic alterations and TKI administration, may contribute to the limited efficacy of immunotherapy observed in EGFR-mutant patients and patients who have received TKI treatment.


Subject(s)
B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Extracellular Vesicles , Lung Neoplasms , Mutation , Protein Kinase Inhibitors , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Female , Cell Line, Tumor , Male , Middle Aged , Aged , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Immunosuppression Therapy
2.
Cancer Sci ; 112(6): 2522-2532, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33728806

ABSTRACT

The 2019 novel coronavirus has spread rapidly around the world. Cancer patients seem to be more susceptible to infection and disease deterioration, but the factors affecting the deterioration remain unclear. We aimed to develop an individualized model for prediction of coronavirus disease (COVID-19) deterioration in cancer patients. The clinical data of 276 cancer patients diagnosed with COVID-19 in 33 designated hospitals of Hubei, China from December 21, 2019 to March 18, 2020, were collected and randomly divided into a training and a validation cohort by a ratio of 2:1. Cox stepwise regression analysis was carried out to select prognostic factors. The prediction model was developed in the training cohort. The predictive accuracy of the model was quantified by C-index and time-dependent area under the receiver operating characteristic curve (t-AUC). Internal validation was assessed by the validation cohort. Risk stratification based on the model was carried out. Decision curve analysis (DCA) were used to evaluate the clinical usefulness of the model. We found age, cancer type, computed tomography baseline image features (ground glass opacity and consolidation), laboratory findings (lymphocyte count, serum levels of C-reactive protein, aspartate aminotransferase, direct bilirubin, urea, and d-dimer) were significantly associated with symptomatic deterioration. The C-index of the model was 0.755 in the training cohort and 0.779 in the validation cohort. The t-AUC values were above 0.7 within 8 weeks both in the training and validation cohorts. Patients were divided into two risk groups based on the nomogram: low-risk (total points ≤ 9.98) and high-risk (total points > 9.98) group. The Kaplan-Meier deterioration-free survival of COVID-19 curves presented significant discrimination between the two risk groups in both training and validation cohorts. The model indicated good clinical applicability by DCA curves. This study presents an individualized nomogram model to individually predict the possibility of symptomatic deterioration of COVID-19 in patients with cancer.


Subject(s)
COVID-19/mortality , Neoplasms/virology , Nomograms , Aged , Area Under Curve , China , Decision Support Techniques , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasms/mortality , Precision Medicine , Retrospective Studies , Risk Factors , Survival Analysis
3.
World J Gastrointest Oncol ; 12(1): 101-112, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31966918

ABSTRACT

BACKGROUND: Primary gastric adenosquamous carcinoma (ASC) is an exceedingly rare histological subtype. Gastric signet ring cell carcinoma (SRC) is a unique subtype with distinct tumor biology and clinical features. The prognosis of gastric ASC vs SRC has not been well established to date. We hypothesized that further knowledge about these distinct cancers would improve the clinical management of such patients. AIM: To investigate the clinicopathological characteristics and prognosis of gastric ASC vs SRC. METHODS: A cohort of gastric cancer patients was retrospectively collected from the Surveillance, epidemiology, and end results program database. The 1:4 propensity score matching was performed among this cohort. The clinicopathological features and prognosis of gastric ASC were compared with gastric SRC by descriptive statistics. Kaplan-Meier method was utilized to calculate the median survival of the two groups of patients. Cox proportional hazard regression models were used to identify prognostic factors. RESULTS: Totally 6063 patients with gastric ASC or SRC were identified. A cohort of 465 patients was recruited to the matched population, including 370 patients with SRC and 95 patients with ASC. Gastric ASC showed an inferior prognosis to SRC after propensity score matching. In the post-matching cohort, the median cancer specific survival was 13.0 (9.7-16.3) mo in the ASC group vs 20.0 (15.7-24.3) mo in the SRC group, and the median overall survival had a similar trend (P < 0.05). ASC and higher tumor-node-metastasis stage were independently associated with a poor survival, while radiotherapy and surgery were independent protective factors for improved prognosis. Subgroup survival analysis revealed that the prognosis of ASC was inferior to SRC only in stages I and II patients. CONCLUSION: ASC may have an inferior prognosis to SRC in patients with stages I and II gastric cancer. Our study supports radiotherapy and surgery for the future management of this clinically rare entity.

4.
Thorac Cancer ; 10(5): 1136-1140, 2019 05.
Article in English | MEDLINE | ID: mdl-30957423

ABSTRACT

BACKGROUND: This study was conducted to investigate if radiotherapy improved the overall survival (OS) of patients with oligometastatic non-small cell lung cancer (NSCLC). METHODS: From January 2012 to August 2015, 323 NSCLC patients with distant metastasis were administered radiotherapy. Ninety-five patients with oligometastatic NSCLC who were sensitive to the initial chemotherapy were treated with radiotherapy for the residual lesions. Initial treatment consisted of four to six cycles of induction chemotherapy. If the patients responded to the initial treatment without developing new metastases, the residual sites were radiated at a total dose of 56-66 Gy, including the primary and metastatic sites. OS, progression-free survival, and sites of progression were assessed. The Kaplan-Meier method was used to estimate the OS and progression-free survival probabilities. RESULTS: The median survival of the whole cohort was 15 months (95% confidence interval 6-40) and the median time to progression was 11 months (95% confidence interval 4-24). Sixty-seven patients had died by the end of follow-up. The one-year and two-year OS rates were 58% and 23%, respectively. Patients progressed either with brain (n = 14), bone (n = 11), lung (n = 10), liver (n = 7), adrenal gland (n = 5), or seven other sites of metastases (n = 3). Acute grade III esophageal toxicity was observed in 17 patients (18%) and grade III pulmonary toxicity in seven patients (7%). CONCLUSION: Oligometastatic non-progressive NSCLC patients may benefit from aggressive radiotherapy to the residual lesions with acceptable toxicity after systemic chemotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms/radiotherapy , Lung Neoplasms/secondary , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/mortality , Combined Modality Therapy , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Grading , Prognosis , Recurrence , Treatment Outcome , Tumor Burden
5.
Oncol Lett ; 10(6): 3613-3618, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26788178

ABSTRACT

The aim of the present study was to investigate the acceleration of pulmonary metastasis due to pulmonary injury caused by radiation treatment in a mouse model of breast cancer, in addition to determining the associated mechanism. The passive metastatic breast cancer model was used in radiation-treated BALB/c mice. In total, 24 mice were randomly separated into two groups, with 12 mice per group, and the groups were treated with or without pulmonary radiation. The survival time and variation of the weights of the lungs, spleen and liver were recorded. Lung metastasis was also evaluated, and chemokine (C-X-C motif) ligand 12 (CXCL12)/chemokine (C-X-C motif) receptor 4 (CXCR4) expression was determined. The results revealed that the group with radiation-induced pulmonary injury exhibited an increased incidence of pulmonary metastasis and shorter survival time compared with the mice without pulmonary radiation. The radiation-treated group possessed an increased number of metastatic nodules in the lungs, but metastasis was not evident in the liver and spleen. The CXCL12/CXCR4 axis was markedly expressed and the expression was significantly increased subsequent to radiation compared with the expression in normal lung tissues. The present study demonstrated that radiation-induced pulmonary injury may accelerate metastatic tumor growth and decrease the overall survival rate of the mice following in situ injection of tumor cells. Tumor localization and growth may have been favored by metastatic conditioning in the lung subsequent to radiotherapy. The CXCL12/CXCR4 axis may affect key elements in the multistep process of metastasis induced by radiation injury.

6.
World J Gastrointest Oncol ; 6(12): 444-9, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25516778

ABSTRACT

AIM: To evaluate the potential prognostic value of GNAS1 T393C polymorphism in advanced non-small cell lung cancer. METHODS: We extracted genomic DNA from the peripheral blood leucocytes of 94 patients with advanced non-small cell lung cancer. Quantitative real-time polymerase chain reaction was used to determine the allelic discrimination. The correlation between genotype and overall survival was evaluated using the multivariate analysis and Kaplan-Meier approach. RESULTS: Thirty-eight out of 94 (40%) patients displayed a TT genotype, 29 out of 94 (31%) a CT genotype and 27 out of 94 (29%) a CC genotype. The median survival of TT (25 mo) genotype carriers was longer than CT (12 mo) or CC (8 mo) genotype carriers. The favorable TT genotype predicted better overall survival (OS) (2-year OS: 48%; P =0.01) compared with CT (2-year OS: 18%) or CC (2-year OS: 15%) genotype. However, dichotomization between C-genotypes (CC + CT) and T-genotypes (TT) revealed significantly lower survival rates (2-year OS: 16%; P = 0.01) for C allele carriers. CONCLUSION: Our data provided strong evidence that the GNAS1 T393C genetic polymorphism influenced the prognosis in advanced non-small lung cancer with a worse outcome for C allele carriers.

SELECTION OF CITATIONS
SEARCH DETAIL