1
|
Xu Y, Ma D, Qin Y, Liu H. Prognostic significance of pathological response and lymph node status in neoadjuvant immunotherapy for potentially resectable non-small cell lung cancer. Ann Med 2025; 57:2453825. [PMID: 39838946 PMCID: PMC11755730 DOI: 10.1080/07853890.2025.2453825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE To evaluate the predictive value of pathological response and lymph node status on progression-free survival (PFS) in patients with potentially resectable non-small cell lung cancer (NSCLC) receiving neoadjuvant immunotherapy. METHODS A retrospective analysis was conducted on 143 patients with potentially resectable NSCLC who underwent neoadjuvant immunotherapy followed by surgical resection. Pathological response, lymph node involvement, and clinical outcomes were comprehensively assessed using Kaplan-Meier analysis and Cox regression. RESULTS Both major pathological response (MPR) and complete pathological response (CPR) significantly correlated with improved PFS (p < .01), with no statistically significant difference between them (p = .15). Lymph node involvement adversely affected PFS (p < .01). A novel risk stratification approach based on pathological response and nodal status effectively distinguished prognostic groups, with 3-year PFS rates of 98.9%, 78.9%, and 53.3%. Cox regression identified gender (HR = 0.25, p = .03), pathological response (HR = 6.02, p < .01), and lymph node stage (HR = 2.30, p = .01) as independent PFS predictors. CONCLUSION In potentially resectable NSCLC, MPR and CPR demonstrate similar PFS benefits after neoadjuvant immunotherapy. Lymph node status significantly influences prognosis, even in initially unresectable cases. The proposed risk stratification provides a valuable tool for personalized management in this challenging patient population.
Collapse
Affiliation(s)
- Yuan Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Dongjie Ma
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yingzhi Qin
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Hongsheng Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Jiang M, Zhang K, Wei G, Qi F, Yu D, Ma J, Zhang X, Chen L, Xie Y, Yu Z, Chen J, Chen D. HDAC4 super-enhancer drives CEBPB-mediated TWIST2 transcription to promote chemoresistance in LUAD. Cancer Lett 2025; 623:217716. [PMID: 40222483 DOI: 10.1016/j.canlet.2025.217716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Lung cancer remains one of the most prevalent malignancies worldwide. This study investigates the role of histone deacetylase 4 (HDAC4) in mediating chemoresistance in lung adenocarcinoma (LUAD). Super-enhancers (SEs), known to regulate aberrant gene expression, are critical drivers of tumor progression. We identified a specific super-enhancer region associated with HDAC4, referred to as HDAC4-SE. Among its nearby genes, TWIST2 emerged as a key player, strongly linked to chemoresistance and the epithelial-to-mesenchymal transition (EMT). We demonstrated that HDAC4-SE regulates TWIST2 expression, thereby contributing to chemoresistance in LUAD. Through bioinformatics analysis, we identified transcription factors binding to both the promoter of TWIST2 and the activation region of HDAC4-SE, with CCAAT/enhancer-binding protein beta (CEBPB) identified as a central regulator. Chromatin immunoprecipitation (ChIP) assays confirmed that CEBPB binds to both the HDAC4-SE and the TWIST2 promoter. Additionally, our investigation into the involvement of long non-coding RNAs (lncRNAs) revealed that LINC01940 might mediate the regulatory effects of HDAC4-SE on downstream genes. In conclusion, we uncovered a novel HDAC4-SE/LINC01940/CEBPB/TWIST2 signaling pathway that drives chemoresistance and tumor progression in LUAD. This pathway offers promising insights into potential therapeutic targets to overcome chemoresistance in lung cancer.
Collapse
Affiliation(s)
- Min Jiang
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, No.188 Shizi Street, Gusu District, Suzhou, 215006, Jiangsu, China
| | - Kai Zhang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Guohao Wei
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, 210003, Nanjing, China
| | - Feng Qi
- Department of Pharmacy, the Yancheng Clinical College of Xuzhou Medical University, the First People's Hospital of Yancheng, No.166 West Yulong Road, Yancheng, 224006, Jiangsu, China
| | - Danlei Yu
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, No.188 Shizi Street, Gusu District, Suzhou, 215006, Jiangsu, China
| | - Jingjing Ma
- Department of Pharmacy, Dushu Lake Hospital, Soochow University, No.9 Chongwen Road, Suzhou, 215100, Jiangsu, China
| | - Xiaofei Zhang
- Department of Medical Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No.160 Pujian Road, Pudong New District, Shanghai, 200127, China
| | - Longbang Chen
- Department of Medical Oncology, Nanjing Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Yuhao Xie
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, 11439, USA
| | - Zhengyuan Yu
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, No.188 Shizi Street, Gusu District, Suzhou, 215006, Jiangsu, China.
| | - Jing Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, No.138 Xianlin Avenue, Nanjing, 210023, Jiangsu, China; Jiangsu Province Engineering Research Center of TCM Health Preservation, Nanjing, Jiangsu, China.
| | - Dongqin Chen
- Department of Medical Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No.160 Pujian Road, Pudong New District, Shanghai, 200127, China; Department of Oncology, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, No. 666 Shengli Road, Nantong, 226000, Jiangsu, China; Department of Medical Oncology, Baoshan Branch, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No.1058 Huanzhen North Road, Baoshan District, Shanghai, 200444, China.
| |
Collapse
|
3
|
Sun R, Li Y, Feng Y, Shao X, Li R, Li H, Sun S, Wang J. PFN1 Knockdown Aggravates Mitophagy to Retard Lung Adenocarcinoma Initiation and M2 Macrophage Polarization. Mol Biotechnol 2025; 67:2673-2684. [PMID: 39120820 DOI: 10.1007/s12033-024-01228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/17/2024] [Indexed: 08/10/2024]
Abstract
Tumor-associated macrophages (TAM) are considered as crucial influencing factors of lung adenocarcinoma (LUAD) carcinogenesis and metastasis. Profilin 1 (PFN1) has been proposed as a potent driver of migration and drug resistance in LUAD. The focus of this work was to figure out the functional mechanism of PFN1 in macrophage polarization in LUAD. PFN1 expression and its significance in patients' survival were detected by ENCORI and Kaplan-Meier Plotter. RT-qPCR and western blotting examined PFN1 expression in LUAD cells. CCK-8 assay and colony formation assay detected cell proliferation. Flow cytometry detected cell apoptosis. Relevant assay kit tested caspase3 concentration. Western blotting analyzed the expression of proliferation- and apoptosis-related proteins. RT-qPCR and immunofluorescence staining measured M1 and M2 macrophages markers. Mitophagy was assessed by MitoTracker Red staining, immunofluorescence staining, and western blotting. PFN1 expression was increased in LUAD tissues and cells and correlated with the poor survival rate of LUAD patients. Deficiency of PFN1 hindered the proliferation, whereas facilitated the apoptosis of LUAD cells. Additionally, PFN1 interference impaired M2 macrophage polarization. Moreover, PFN1 knockdown exacerbated the mitophagy in LUAD cells and mitophagy inhibitor mitochondrial division inhibitor 1 (Mdivi-1) notably reversed the effects of PFN1 down-regulation on the proliferation, apoptosis as well as macrophage polarization in LUAD cells. To sum up, activation of mitophagy initiated by PFN1 depletion might obstruct the occurrence and M2 macrophage polarization in LUAD.
Collapse
Affiliation(s)
- Rongrong Sun
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China.
| | - Yang Li
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China
| | - Yu Feng
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China
| | - Xiaoyan Shao
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China
| | - Rantian Li
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China
| | - Hao Li
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China
| | - Sanyuan Sun
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China
| | - Jiangbo Wang
- Department of Oncology, Xuzhou Central Hospital, 199 Jiefang South Road, XuZhou, 221000, Jiangsu, China.
| |
Collapse
|
4
|
Wang R, Li X, Long L, Li M, Chen H, Zhang H, Ruan W, Zhang H, Zheng P, Xu S. Novel ALK inhibitors containing DAAP fragments: Rational drug design and anti-tumor activity research. Bioorg Chem 2025; 160:108416. [PMID: 40209355 DOI: 10.1016/j.bioorg.2025.108416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase belonging to the insulin receptor subfamily, involved in cellular signaling pathways associated with insulin. While ALK gene remains inactive in normal human tissues, it exhibits high expression specifically in the central nervous system. However, dysregulation of the ALK gene has been implicated in non-small cell lung cancer (NSCLC) development. Although commercially available ALK inhibitors demonstrate favorable clinical efficacy against most Ceritinib-resistant mutants, they exhibit resistance towards G1202R mutants. Therefore, developing novel ALK inhibitors is crucial for addressing drug resistance in patients. We designed and synthesized 48 novel 2,4-diarylpyrimidine-based ALK inhibitors and investigated their antitumor activities. Among them, Ld-10 showed significant inhibitory activity against ALK kinase with an IC50 value of 1135 nM and demonstrated excellent antiproliferative activity against lung cancer cells H2228 with an IC50 value of 1.35 ± 0.13 μM. To further validate the antitumor potential of Ld-10, we conducted a series of in vitro pharmacological experiments. These included a hemolysis assay to confirm its low toxicity profile, an AO assay, a JC-1 staining assay, and a Calcein-AM/PI cell double staining assay for assessing apoptosis induction. Additionally, we performed dose-dependent arrest at G0/G1 phase to evaluate inhibition of cell growth and carried out cell cycle analysis and cloning experiments to provide evidence for significant tumor growth inhibition by compound Ld-10. In vivo pharmacological experiments demonstrated effective tumor growth inhibition without any significant toxic effects on mouse organs caused by Ld-10 administration. Based on these comprehensive findings from our experimental investigations, it can be concluded that Ld-10 holds promising potential as a novel ALK inhibitor.
Collapse
Affiliation(s)
- Ran Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China
| | - Xiangjing Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China
| | - Li Long
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China
| | - Min Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China
| | - Huijing Chen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China
| | - Han Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China
| | - Wei Ruan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China
| | - Hong Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China.
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China.
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, 330013, China.
| |
Collapse
|
5
|
Chen X, Wang Y, Liu Z, He Y, Gao H, Guan X, Chen L, Chen S, Xing X, Chen W, Li D, Wang Q. Role of the USP7/FOXO3A axis in environmentally relevant doses of arsenic-induced lung carcinogenesis: Insights from bioinformatics analysis and model of human epithelial cell malignant transformation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118224. [PMID: 40273609 DOI: 10.1016/j.ecoenv.2025.118224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/05/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025]
Abstract
Arsenic (As) is classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC). Exposure to As has been associated with an increased risk of various cancers, particularly lung cancer. However, the precise molecular mechanisms contributing to this carcinogenesis are not well understood. In our study, we analyzed transcriptomic data from the GEO database (GSE36684), identifying 764 differentially expressed genes (DEGs) in BEAS-2B cells treated with environmentally relevant doses of As for 8 weeks. A KEGG pathway enrichment analysis suggested that the FoxO pathway activation might be a novel key signaling event in As-induced carcinogenesis. We further analyzed the expression of 11 DEGs involved in the FoxO pathway using the TCGA-LUSC dataset. The findings revealed that four genes displayed expression patterns in tumor tissues consistent with those observed after As treatment in GEO dataset. Among them, USP7 was upregulated, while ATM, S1PR1, and PLK2 were downregulated in cancer tissues. High USP7 expression was specifically linked to a poor prognosis in lung squamous cell carcinoma (LUSC). To explore the role of USP7 in As-induced malignant transformation, BEAS-2B cells were exposed to NaAsO2 concentrations of 0.2 μM and 2 μM for up to 20 weeks. Experimental results confirmed that NaAsO2 treatment suppressed the FoxO transcriptional activity by upregulating USP7 expression, subsequently downregulating ATM and PLK2 expression, which led to abnormalities in cell cycle regulation and apoptosis. Notably, knocking down USP7 in As-transformed cells resulted in significant reductions in cell proliferation, colony formation, and tumor formation ability in nude mice, indicating the USP7-regulated FOXO3A pathway could be central to As-induced lung carcinogenesis. Moreover, our research demonstrated that USP7 inhibited FOXO3A's ability to translocate from the cytoplasm to the nucleus by affecting its monoubiquitination status. Additionally, we speculated that As-induced the elevation of USP7 expression due to the excessive inflammatory cytokines secretion and the activation of mTORC1/WTAP pathway. These findings offer novel insights into the molecular mechanisms underlying As-mediated lung cancer.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuqing Wang
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ziqi Liu
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yujie He
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Huan Gao
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xinchao Guan
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shen Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiumei Xing
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Qing Wang
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
6
|
Zhang G, Lu S, Ren Z, Wei L, Chen C, Tao P, Pan X. SIRT2 as a Potential Biomarker in Lung Adenocarcinoma: Implications for Immune Infiltration. Mol Biotechnol 2025; 67:2305-2318. [PMID: 38902578 DOI: 10.1007/s12033-024-01198-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/13/2024] [Indexed: 06/22/2024]
Abstract
SIRT2 play important roles in cell cycle and cellular metabolism in the development of non-small cell lung cancer (NSCLC), and SIRT2 exhibits its therapeutic effect on NSCLC tumors with high expression of SIRT2. Nevertheless, the clinical relevance of SIRT2 in lung adenocarcinoma (LUAD), particularly its impact on tumor growth and prognostic implications, remains obscure. This investigation entailed a comprehensive analysis of SIRT2 mRNA and protein expression levels in diverse tumor and corresponding healthy tissues, utilizing databases such as TIMER 2.0, UALCAN, and HPA. Prognostic correlations of SIRT2 expression in LUAD patients, stratified by distinct clinicopathological characteristics, were evaluated using the KM Plotter database. Additionally, the TCGA and TIMER 2.0 databases were employed to assess the relationship between SIRT2 and immune infiltration, as well as to calculate immunity, stromal, and estimation scores, thus elucidating the role of SIRT2 in modulating tumor immunotherapy responses. Furthermore, Gene Set Enrichment Analysis (GSEA) was utilized to elucidate SIRT2's biological functions in pan-cancer cells. Our findings revealed a marked reduction in both mRNA and protein levels of SIRT2 in LUAD tumors relative to healthy tissue. Survival analysis indicated that diminished SIRT2 expression correlates with adverse prognostic outcomes in LUAD. Furthermore, SIRT2 expression demonstrated a significant association with various clinicopathologic attributes of LUAD patients, influencing survival outcomes across different clinicopathologic states. Functional enrichment analyses highlighted SIRT2's involvement in cell cycle regulation and immune response. Notably, SIRT2 exhibited a positive correlation with immune cell infiltration, including natural killer (NK) cells, macrophages, and dendritic cells (DCs). In summary, SIRT2 was a potential prognostic biomarker for LUAD and and a new immunotherapy target.
Collapse
Affiliation(s)
- Guining Zhang
- Department of Scientific Research, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Shuyu Lu
- Department of Anaesthesia, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning, 530007, Guangxi, China
| | - Zhiling Ren
- Department of Mental Health, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Lijuan Wei
- Graduate School, Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Chunxi Chen
- Graduate School, Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Pinyue Tao
- Department of Anaesthesia, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning, 530007, Guangxi, China.
| | - Xiao Pan
- The Second Ward of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning, 530007, Guangxi, China.
| |
Collapse
|
7
|
Su XE, Lin CL, Wang HG, Jing-Liu, Peng CB, He HF, Wu S, Huang XF, Lin S, Xie BY. Development and Validation of a Machine Learning-Based Predictive Model for Postoperative Frailty in Patients with Non-Small Cell Lung Cancer and Its Relation to Early Recovery. Ann Surg Oncol 2025:10.1245/s10434-025-17353-2. [PMID: 40366574 DOI: 10.1245/s10434-025-17353-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE This study was designed to evaluate the postoperative frailty status of patients with non-small cell lung cancer, identify influencing factors, establish a machine learning-based prediction model, and explore the correlation between frailty status at 3 months and early recovery at 1 month postoperatively. METHODS This retrospective analysis included patients with non-small cell lung cancer who underwent surgery at our hospital from 2021 to 2024. Clinical variables, including demographics, tumor characteristics, treatment, and laboratory tests, were analyzed. Feature selection and model construction were performed by using LASSO regression. Cross-validation assessed the accuracy of the models. Frailty at 3 months and quality of recovery at 1 month postoperatively were measured by using the Tilburg Frailty Index and Quality of Recovery (QoR-15) scales, respectively. RESULTS A total of 1,013 patients were included. The initial model achieved an AUC of 0.833, accuracy of 0.854, recall of 0.382, and F1 score of 0.502 in the training set, and an AUC of 0.786, accuracy of 0.857, recall of 0.242, and F1 score of 0.364 in the validation set. Of the patients, 190 (18.8%) developed frailty at 3 months postoperatively. After applying Synthetic Minority oversampling Technique to balance the data, the model's performance improved (area under the curve [AUC] 0.850, accuracy 0.791, recall 0.818, and F1 score 0.795 for the training set; AUC 0.819, accuracy 0.778, recall 0.762, and F1 score 0.781 for the test set). Additionally, we developed a nomogram to visually represent the predictive model, enabling clinicians to easily assess frailty risk in individuals based on key factors. Correlation analyses showed that frailty at 3 months was moderately negatively correlated with early recovery at 1 month (correlation coefficient = - 0.370). CONCLUSIONS This study developed a predictive model of postsurgical frailty in lung cancer, providing insights into personalized patient management and early recovery improvement. Further studies should explore the clinical application of the model.
Collapse
Affiliation(s)
- Xue-E Su
- Department of Anaesthesia, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Cui-Liu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Huai-Gang Wang
- Neusoft Research of Intelligent Healthcare Technology, Co. Ltd., Shenyang City, Liaoning Province, China
| | - Jing-Liu
- Neusoft Research of Intelligent Healthcare Technology, Co. Ltd., Shenyang City, Liaoning Province, China
| | - Cheng-Bao Peng
- Neusoft Research of Intelligent Healthcare Technology, Co. Ltd., Shenyang City, Liaoning Province, China
| | - He-Fan He
- Department of Anaesthesia, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shanhu Wu
- Department of Anaesthesia, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, Wollongong, Australia
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| | - Bao-Yuan Xie
- Department of Nursing, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| |
Collapse
|
8
|
Li H, Li S, Kanamori Y, Liu S, Moroishi T. Auranofin resensitizes ferroptosis-resistant lung cancer cells to ferroptosis inducers. Biochem Biophys Res Commun 2025; 770:151992. [PMID: 40373379 DOI: 10.1016/j.bbrc.2025.151992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/23/2025] [Accepted: 05/09/2025] [Indexed: 05/17/2025]
Abstract
Lung cancer, a major cause of cancer-related mortality, has limited therapeutic options, especially for advanced cases. Ferroptosis, an iron-dependent form of cell death, is a potential therapeutic strategy for this disease; however, resistance mechanisms in the tumor microenvironment impede its effectiveness. Therefore, in this study, we aimed to investigate the efficacy of sulfasalazine (SAS), a ferroptosis inducer, and auranofin (AUR), a Food and Drug Administration-approved anti-inflammatory agent, combination to counteract ferroptosis resistance in lung cancer. SAS induced ferroptosis in vitro; however, its efficacy in vivo was limited, possibly because of factors, such as nutrient deprivation and high cell density, in the microenvironment that suppressed the activities of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of ferroptosis resistance. Screening of 2483 drugs revealed AUR as a compound resensitizing the YAP/TAZ-deficient lung cancer cells to ferroptosis. Moreover, SAS and AUR combination significantly enhanced lipid peroxidation and reactive oxygen species accumulation, further driving ferroptosis in cells. This combination effectively inhibited tumor growth and enhanced survival in a murine lung cancer model. Overall, our findings suggest that AUR potentiates ferroptosis-based therapies, serving as an effective candidate to overcome ferroptosis resistance in lung cancer.
Collapse
Affiliation(s)
- Hao Li
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Shuran Li
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yohei Kanamori
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Saisai Liu
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan; Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan; Division of Cellular Dynamics, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Tokyo, 113-8510, Japan.
| |
Collapse
|
9
|
Li S, Huang Z, Zhong X, Zhou Y, Jiang H. The real-world safety profile of pemetrexed and platinum with or without pembrolizumab: insights from a comparative analysis of FAERS database. BMC Cancer 2025; 25:767. [PMID: 40275143 PMCID: PMC12020046 DOI: 10.1186/s12885-025-14171-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Lung cancer (LC) is a leading cause of cancer-related mortality worldwide. The combination of immune checkpoint inhibitors (ICIs) with chemotherapy significantly extends survival but increases the risk of treatment-related toxicity. To explore the impact of adding pembrolizumab to pemetrexed and platinum on treatment-related toxicity, this study utilized the FDA Adverse Event Reporting System (FAERS) to assess the safety of pemetrexed and platinum with or without pembrolizumab in LC patients. METHODS We collected data from FAERS database between the second quarter of 2017 and the third quarter of 2024. Disproportionality analysis was conducted using the Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-Item Gamma Poisson Shrinker (MGPS). Additionally, comparative analysis was performed using the ROR method. RESULTS Among LC patients receiving chemotherapy alone (pemetrexed + platinum) and combination therapy (pembrolizumab + pemetrexed + platinum), adverse event (AE) reports were 2871 and 5443 cases, respectively. Compared to chemotherapy alone, combination therapy was associated with a higher risk of renal and urinary disorders, hepatobiliary disorders, and interstitial lung disease (ILD), pneumonitis and other AEs. Subgroup analysis revealed that gender and age may be influential factors in the occurrence of AEs. Combination therapy prolonged the time to onset of AEs. CONCLUSIONS In the real world, combination therapy increases the risk of certain AEs, particularly in specific patient subgroups. These findings emphasize the importance of personalized treatment strategies and AE monitoring, particularly during the first three months of therapy.
Collapse
Affiliation(s)
- Shun Li
- Department of Radiation Oncology, The first affiliated hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China
- Joint Research Center for Regional Diseases of IHM, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Zhifei Huang
- Department of Radiation Oncology, The first affiliated hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China
- Joint Research Center for Regional Diseases of IHM, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Xiaoyu Zhong
- Department of Radiation Oncology, The first affiliated hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China
- Joint Research Center for Regional Diseases of IHM, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Yan Zhou
- Department of Radiation Oncology, The first affiliated hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China.
- Joint Research Center for Regional Diseases of IHM, Bengbu Medical University, Bengbu, Anhui, 233030, China.
| | - Hao Jiang
- Department of Radiation Oncology, The first affiliated hospital of Bengbu Medical University, Bengbu, Anhui, 233000, China.
- Joint Research Center for Regional Diseases of IHM, Bengbu Medical University, Bengbu, Anhui, 233030, China.
| |
Collapse
|
10
|
Rong T, Ai C, Yang T, Wu Q, Zhang M. Clinical features and prognostic nomogram development for cancer-specific death in patients with dual primary lung cancer: a population-based study from SEER database. J Cardiothorac Surg 2025; 20:190. [PMID: 40217288 PMCID: PMC11992714 DOI: 10.1186/s13019-025-03385-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 03/09/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE This study aimed to develop a concise and valid clinical prediction model to assess the survival prognostic risk of cancer-specific death in patients with dual primary lung cancer (DPLC). DATA SOURCE Surveillance, epidemiology, and end results (SEER) database. DESIGN A retrospective population-based study. METHODS Data of DPLC patients from the database from 1992 to 2020 were collected. The number of DPLC patients was determined based on the first primary LC (FPLC) and second primary LC (SPLC), and patients were randomly assigned to a training set and a testing set in a 7:3 ratio. The primary endpoint was cancer-specific survival (CSS). Kaplan-Meier survival analysis was performed to construct survival curves. Cox analysis and bilateral stepwise regression were used to analyze prognostic factors for cancer-specific death in patients and establish the nomogram. The discriminative ability of the nomogram was assayed by C-index and calibration curves, decision-making ability was assessed by decision curve analysis (DCA), and nomogram performance was measured by receiver operating characteristic (ROC) curves. RESULTS This study included 997 DPLC patients, divided into a training set (n = 698) and a testing set (n = 299) in a 7:3 ratio. Age, gender, histological type, surgery, chemotherapy, T stage, N stage, and tumor size were identified as risk factors affecting CSS in DPLC patients (P < 0.05) and were utilized to establish a nomogram. The C-index of the nomogram in the training set was 0.671, and the AUC values of ROC curves for 1-year, 3-year, and 5-year survival rates were 0.84, 0.78, and 0.74, respectively. The C-index of the testing set was 0.644, and the AUC values were 0.72, 0.74, and 0.75, respectively. Calibration curves for both sets were close to the diagonal line, indicating good predictive ability of the nomogram. DCA curves demonstrated the good decision-making ability of the nomogram. CONCLUSION This study revealed the clinical features of DPLC patients and developed an effective nomogram for predicting CSS, which can assist clinicians in making accurate and personalized clinical decisions regarding patient treatment.
Collapse
Affiliation(s)
- Tenghao Rong
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing Medical University, No. 9, Shuangxing Avenue, Bishan District, Chongqing, 402760, China
| | - Cheng Ai
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing Medical University, No. 9, Shuangxing Avenue, Bishan District, Chongqing, 402760, China
| | - Tong Yang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Qingchen Wu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuzhong District, Chongqing, 400016, China.
| | - Min Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
11
|
Birk S, Bonafonte-Pardàs I, Feriz AM, Boxall A, Agirre E, Memi F, Maguza A, Yadav A, Armingol E, Fan R, Castelo-Branco G, Theis FJ, Bayraktar OA, Talavera-López C, Lotfollahi M. Quantitative characterization of cell niches in spatially resolved omics data. Nat Genet 2025; 57:897-909. [PMID: 40102688 PMCID: PMC11985353 DOI: 10.1038/s41588-025-02120-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025]
Abstract
Spatial omics enable the characterization of colocalized cell communities that coordinate specific functions within tissues. These communities, or niches, are shaped by interactions between neighboring cells, yet existing computational methods rarely leverage such interactions for their identification and characterization. To address this gap, here we introduce NicheCompass, a graph deep-learning method that models cellular communication to learn interpretable cell embeddings that encode signaling events, enabling the identification of niches and their underlying processes. Unlike existing methods, NicheCompass quantitatively characterizes niches based on communication pathways and consistently outperforms alternatives. We show its versatility by mapping tissue architecture during mouse embryonic development and delineating tumor niches in human cancers, including a spatial reference mapping application. Finally, we extend its capabilities to spatial multi-omics, demonstrate cross-technology integration with datasets from different sequencing platforms and construct a whole mouse brain spatial atlas comprising 8.4 million cells, highlighting NicheCompass' scalability. Overall, NicheCompass provides a scalable framework for identifying and analyzing niches through signaling events.
Collapse
Affiliation(s)
- Sebastian Birk
- Institute of AI for Health, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- Würzburg Institute of Systems Immunology (WüSI), University of Würzburg, Würzburg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Irene Bonafonte-Pardàs
- Institute of Computational Biology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
- Biomedical Center (BMC), Physiological Chemistry, Faculty of Medicine, Ludwig Maximilian University of Munich, Planegg-Martinsried, Germany
| | | | - Adam Boxall
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fani Memi
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Anna Maguza
- Würzburg Institute of Systems Immunology (WüSI), University of Würzburg, Würzburg, Germany
- Faculty of Medicine, University of Würzburg, Würzburg, Germany
| | - Anamika Yadav
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Erick Armingol
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Human and Translational Immunology Program, Yale University School of Medicine, New Haven, CT, USA
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden
| | - Fabian J Theis
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Institute of Computational Biology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | | | - Carlos Talavera-López
- Würzburg Institute of Systems Immunology (WüSI), University of Würzburg, Würzburg, Germany.
- Faculty of Medicine, University of Würzburg, Würzburg, Germany.
| | - Mohammad Lotfollahi
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Institute of Computational Biology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
12
|
Heng Y, Wang F, Zhang Z, Lin Z, Zhao D, Li Q. PRMT7 Inhibitor SGC3027 Enhances Radiotherapy Efficacy via Activating ATM Kinase in Non-Small Cell Lung Carcinoma. Radiat Res 2025; 203:284-292. [PMID: 40015317 DOI: 10.1667/rade-24-00242.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of tumor-related death in humans. Radiotherapy is a crucial strategy for NSCLC treatment, although its effectiveness is limited by the radio-resistance of tumor cells. Our current research finds that the protein arginine methyltransferase 7 (PRMT7) is upregulated in NSCLC and correlates with poor prognosis. Pharmacological inhibition of PRMT7 by SGC3027, a specific small-molecule PRMT7 inhibitor, suppresses the proliferation, migration and invasion of NSCLC. Combining irradiation with SGC3027 strengthens the impact of irradiation on the biological behaviors of NSCLC cells. We also find that SGC3027 specifically activates ATM kinase and its downstream cell cycle checkpoint kinases to enhance radiobiological response in NSCLC. These findings underscore the promising therapeutic potential of PRMT7 inhibitors as well as combining PRMT7 inhibition with irradiation exposure for effective NSCLC therapies.
Collapse
Affiliation(s)
- Ya Heng
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| | - Feifei Wang
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| | - Zhonghui Zhang
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| | - Zebang Lin
- Department of Thoracic Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, P.R. China
| | - Dahai Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei 230601, P.R. China
| | - Qiuling Li
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| |
Collapse
|
13
|
Liu C, Lin M, Xin Y, Sun M, Cui J, Liu D, Zang D, Chen J. Metagenomics and Non-Targeted Metabolomics Reveal the Role of Gut Microbiota and Its Metabolites in Brain Metastasis of Non-Small Cell Lung Cancer. Thorac Cancer 2025; 16:e70068. [PMID: 40263747 PMCID: PMC12014518 DOI: 10.1111/1759-7714.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Brain metastasis is a common and severe complication in non-small cell lung cancer (NSCLC) patients, significantly affecting prognosis. However, the role of gut microbiota and its metabolites in NSCLC brain metastasis remains poorly understood. This study aims to explore the relationship between gut microbiota, metabolites, and the development of brain metastasis in NSCLC. METHODS We conducted an integrative analysis combining metagenomics and non-targeted metabolomics on baseline fecal samples from NSCLC patients with brain metastasis (n = 18) and those without distant metastasis (n = 12). Gut microbiota composition and metabolite profiles were detected and analyzed, and statistical methods, including machine learning models, were applied to identify differences and potential biomarkers. RESULTS Significant differences in gut microbiota composition were found between the two groups, with higher microbial diversity observed in patients with brain metastasis. Specific genera, such as Paenibacillus, Fournierella, and Adlercreutzia, were enriched in the brain metastasis group. Metabolomic analysis revealed altered levels of short-chain fatty acids and other metabolites associated with immune modulation and vascular permeability, including angiotensin (1-7). These changes were linked to the metastatic process and may influence brain metastasis development. Furthermore, machine learning models identified key biomarkers, such as Raoultibacter, Mobilibacterium, and N-acetyl-L-glutamic acid, which could serve as valuable indicators for brain metastasis. CONCLUSIONS Our findings suggest that gut microbiota dysbiosis and its metabolic products may contribute to the development of brain metastasis in NSCLC. The identification of microbiota-derived biomarkers holds potential for early detection and therapeutic intervention in NSCLC brain metastasis.
Collapse
Affiliation(s)
- Chen‐Guang Liu
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Mei‐Xi Lin
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Yu Xin
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Man Sun
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Jia Cui
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Dan Liu
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Dan Zang
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Jun Chen
- Department of OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
14
|
Xu L, Ma X, Yang Y, Ding Z, Luo Y. Successful treatment of an elderly patient with lung squamous cell carcinoma by tislelizumab and chemotherapy: a case report with novel imaging findings. Front Immunol 2025; 16:1543114. [PMID: 40196110 PMCID: PMC11973309 DOI: 10.3389/fimmu.2025.1543114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
The advent of immunotherapy has transformed the therapeutic landscape for inoperable, locally advanced Non-Small cell lung cancer (NSCLC), particularly for lung squamous cell carcinoma (LUSC) with a predominance of negative driver genes. Based on the results of clinical trials such as KEYNOTE-042 and KEYNOTE-407, PD-1/PD-L1 inhibitors are now recognized as the standard of care for first-line or second-line treatment in many countries. Among the 17 immune checkpoint inhibitors sanctioned in China, tislelizumab, a domestically developed PD-1 inhibitor, enjoys broad application. Here, we present a case of a patient with LUSC who attained complete remission by cyst formation with the combination of tislelizumab and chemotherapy. Despite the absence of expression data for this patient, imaging studies revealed a reduction in the primary lesion size and the emergence of an uncommon cystic alteration post-treatment with sequential immunochemotherapy and tislelizumab monotherapy. As per the most recent follow-up, the lesion has vanished entirely. This outcome holds significant implications for the treatment of driver gene-negative LUSC by tislelizumab.
Collapse
Affiliation(s)
- Lufan Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinxin Ma
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Yang
- Nantong Tumor Hospital, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Zhiqiang Ding
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Luo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
15
|
He Y, Altuna-Coy A, Acosta-Plasencia M, Molins L, Sánchez-Lorente D, Martinez D, Díaz T, Na R, Marrades RM, Navarro A. Diagnostic and Prognostic Value of hsa_piR_022710, hsa_piR_019822, and hsa_piR_020840 in Early-Stage Non-Small-Cell Lung Cancer: Implications for Recurrence and Survival in Squamous Cell Carcinoma Patients. Int J Mol Sci 2025; 26:2870. [PMID: 40243460 PMCID: PMC11989015 DOI: 10.3390/ijms26072870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Despite significant advancements in early detection and treatment, non-small-cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality. Specifically, in early-stage cases, recurrence after surgery continues to be the principal cause of death for these patients. The urgent need for novel diagnostic and prognostic biomarkers has directed attention towards PIWI-interacting RNAs (piRNAs), a group of small RNAs that regulate genomic stability and epigenetics. Some piRNAs, including hsa_piR_022710, hsa_piR_019822, and hsa_piR_020840, have been described as deregulated in various cancers. This study investigated the expression of these three piRNAs by RT-qPCR in 277 NSCLC patients and developed survival and CART classification models to predict recurrence risk, overall survival (OS), and disease-free survival (DFS). hsa_piR_019822 and hsa_piR_020840 were able to discriminate between tumor and normal tissue, as well as between adenocarcinoma and squamous cell carcinoma (LUSC) patients. Elevated expression of hsa_piR_019822 and hsa_piR_022710 was correlated with an increased risk of recurrence and poorer DFS and OS in LUSC patients. Patients with high hsa_piR_022710 expression more greatly benefited from adjuvant treatment. In summary, higher piRNA levels were associated with an increased risk of recurrence and poorer survival outcomes, especially in LUSC patients, where they may help guide personalized treatment strategies.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/diagnosis
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- Male
- Female
- Lung Neoplasms/genetics
- Lung Neoplasms/diagnosis
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Middle Aged
- Prognosis
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/diagnosis
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/mortality
- Neoplasm Recurrence, Local/genetics
- Biomarkers, Tumor/genetics
- Aged
- RNA, Small Interfering/genetics
- Neoplasm Staging
- Gene Expression Regulation, Neoplastic
- Adult
- Disease-Free Survival
Collapse
Affiliation(s)
- Yangyi He
- Molecular Oncology and Embryology Laboratory, Human Anatomy and Embryology Unit, Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), c. Casanova 143, 08036 Barcelona, Spain; (Y.H.); (A.A.-C.); (M.A.-P.); (T.D.); (R.N.)
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Antonio Altuna-Coy
- Molecular Oncology and Embryology Laboratory, Human Anatomy and Embryology Unit, Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), c. Casanova 143, 08036 Barcelona, Spain; (Y.H.); (A.A.-C.); (M.A.-P.); (T.D.); (R.N.)
| | - Melissa Acosta-Plasencia
- Molecular Oncology and Embryology Laboratory, Human Anatomy and Embryology Unit, Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), c. Casanova 143, 08036 Barcelona, Spain; (Y.H.); (A.A.-C.); (M.A.-P.); (T.D.); (R.N.)
| | - Laureano Molins
- Department of Thoracic Surgery, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036 Barcelona, Spain;
- Thoracic Oncology Unit, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036 Barcelona, Spain; (D.M.); (R.M.M.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c. Villarroel 170, 08036 Barcelona, Spain
| | - David Sánchez-Lorente
- Department of Thoracic Surgery, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, 08202 Sabadell, Spain;
| | - Daniel Martinez
- Thoracic Oncology Unit, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036 Barcelona, Spain; (D.M.); (R.M.M.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c. Villarroel 170, 08036 Barcelona, Spain
- Department of Pathology, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036 Barcelona, Spain
| | - Tania Díaz
- Molecular Oncology and Embryology Laboratory, Human Anatomy and Embryology Unit, Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), c. Casanova 143, 08036 Barcelona, Spain; (Y.H.); (A.A.-C.); (M.A.-P.); (T.D.); (R.N.)
| | - Risha Na
- Molecular Oncology and Embryology Laboratory, Human Anatomy and Embryology Unit, Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), c. Casanova 143, 08036 Barcelona, Spain; (Y.H.); (A.A.-C.); (M.A.-P.); (T.D.); (R.N.)
| | - Ramón M. Marrades
- Thoracic Oncology Unit, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036 Barcelona, Spain; (D.M.); (R.M.M.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c. Villarroel 170, 08036 Barcelona, Spain
- Department of Pneumology, Institut Clínic Respiratori (ICR), Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alfons Navarro
- Molecular Oncology and Embryology Laboratory, Human Anatomy and Embryology Unit, Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, Universitat de Barcelona (UB), c. Casanova 143, 08036 Barcelona, Spain; (Y.H.); (A.A.-C.); (M.A.-P.); (T.D.); (R.N.)
- Thoracic Oncology Unit, Hospital Clínic de Barcelona, Universitat de Barcelona (UB), 08036 Barcelona, Spain; (D.M.); (R.M.M.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c. Villarroel 170, 08036 Barcelona, Spain
| |
Collapse
|
16
|
Chen F, Xiang W, Qiang G. Tanshinone IIA affects the proliferation of A549/Tax by affecting the expression of MMP7 through the PI3K-AKT-mTOR signaling pathway. Discov Oncol 2025; 16:369. [PMID: 40113621 PMCID: PMC11926296 DOI: 10.1007/s12672-025-02152-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE This study aims to explore whether tanshinone IIA can act on paclitaxel-resistant non-small cell lung cancer A549/Tax and analyze the possible mechanisms involved. METHODS Using the Cell Counting Kit-8 (CCK-8), we preliminarily analyzed whether tanshinone IIA has an inhibitory effect on A549/Tax cells. We utilized public datasets, self-collected transcriptome datasets, and drug target analysis to identify potential targets. We employed real-time fluorescent quantitative polymerase chain reaction (RT-qPCR) to detect the expression of core genes before and after drug treatment to analyze potential target genes and validated them using data from The Cancer Genome Atlas (TCGA). We conducted enrichment analysis on co-expressed genes of the target genes to explore potential mechanisms. Furthermore, we employed molecular docking and western blot to verify the possible mechanisms involved. RESULTS The CCK8 results indicated that tanshinone IIA has a significant inhibitory effect on A549/Tax cells. The qPCR results and the analysis of TCGA data indicated that MMP7 is the target gene. Enrichment results of MMP7 co-expressed genes suggested that the PI3K-AKT signaling pathway might play a key role. Molecular docking results indicated that tanshinone IIA has strong binding activity with PI3K, AKT, mTOR, and MMP7. Western blotting results showed that tanshinone IIA might inhibit MMP7 through the PI3K-AKT-mTOR signaling pathway. CONCLUSIONS Tanshinone IIA may affect the proliferation of A549/Tax by influencing the expression of MMP7 through the PI3K-AKT-mTOR signaling pathway.
Collapse
Affiliation(s)
- Fangjun Chen
- Department of Thoracic Surgery, China Japan Friendship Institute of Clinical Medicine Research, Beijing, China
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Wenqiong Xiang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangliang Qiang
- Department of Thoracic Surgery, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
17
|
Luo X, Zhang X, Su D, Li H, Zou M, Xiong Y, Yang L. Deep Clustering-Based Metabolic Stratification of Non-Small Cell Lung Cancer Patients Through Integration of Somatic Mutation Profile and Network Propagation Algorithm. Interdiscip Sci 2025:10.1007/s12539-025-00699-2. [PMID: 40100545 DOI: 10.1007/s12539-025-00699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/20/2025]
Abstract
As a common malignancy of the lower respiratory tract, non-small cell lung cancer (NSCLC) represents a major oncological challenge globally, characterized by high incidence and mortality rates. Recent research highlights the critical involvement of somatic mutations in the onset and development of NSCLC. Stratification of NSCLC patients based on somatic mutation data could facilitate the identification of patients likely to respond to personalized therapeutic strategies. However, stratification of NSCLC patients using somatic mutation data is challenging due to the sparseness of this data. In this study, based on sparse somatic mutation data from 4581 NSCLC patients from the Memorial Sloan Kettering Cancer Center (MSKCC) database, we systematically evaluate the metabolic pathway activity in NSCLC patients through the application of network propagation algorithm and computational biology algorithms. Based on these metabolic pathways associated with prognosis, as recognized through univariate Cox regression analysis, NSCLC patients are stratified using the deep clustering algorithm to explore the optimal classification strategy, thereby establishing biologically meaningful metabolic subtypes of NSCLC patients. The precise NSCLC metabolic subtypes obtained from the network propagation algorithm and deep clustering algorithm are systematically evaluated and validated for survival benefits of immunotherapy. Our research marks progress towards developing a universal approach for classifying NSCLC patients based solely on somatic mutation profiles, employing deep clustering algorithm. The implementation of our research will help to deepen the analysis of NSCLC patients' metabolic subtypes from the perspective of tumor microenvironment, providing a strong basis for the formulation of more precise personalized treatment plans.
Collapse
Affiliation(s)
- Xu Luo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xinpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Dongqing Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Honghao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Min Zou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yuqiang Xiong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
18
|
Zhang J, Wang Y, Wang M, Wang P, Yan H, Wang D, Zheng J, Li B, Chen C, Cao D, Zheng X, Li X, Wang K, Liu Z. Discovery of Novel 2,4,5-Trisubstituted Pyrimidine Derivatives as Potent and Selective FGFR Inhibitors against Gatekeeper Mutants for the Treatment of NSCLC. J Med Chem 2025; 68:5907-5925. [PMID: 40032550 DOI: 10.1021/acs.jmedchem.5c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Fibroblast growth factor receptors (FGFRs) play a critical role in the regulation of cancer cell proliferation, differentiation, and migration. However, the development of acquired resistance to FGFR inhibitors remains a major challenge in treating non-small cell lung cancer (NSCLC), particularly due to mutations at the gatekeeper residue. In this study, we report the discovery of a series of irreversible FGFR inhibitors targeting gatekeeper mutations in FGFR1-3, utilizing a 2,4,5-trisubstituted pyrimidine scaffold. Through rational design, structure-activity relationship optimization, and pharmacokinetic evaluation, compound ng 12l emerged as a promising candidate. It demonstrated potent inhibition of FGFR1-3 gatekeeper mutations in vitro along with favorable pharmacokinetic properties. The efficacy of 12l in targeting FGFR1 gatekeeper mutations was confirmed in assays using L6-FGFR1V561M/F cells. Furthermore, in xenograft models using both H1581 and L6-FGFR1V561M cells, 12l exhibited robust anti-tumor activity with minimal toxicity. These findings position 12l as a promising therapeutic agent for overcoming gatekeeper-mediated resistance in NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Pyrimidines/pharmacology
- Pyrimidines/chemical synthesis
- Pyrimidines/chemistry
- Pyrimidines/therapeutic use
- Pyrimidines/pharmacokinetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Animals
- Structure-Activity Relationship
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/therapeutic use
- Cell Line, Tumor
- Mutation
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/chemical synthesis
- Protein Kinase Inhibitors/therapeutic use
- Mice
- Receptors, Fibroblast Growth Factor/antagonists & inhibitors
- Mice, Nude
- Xenograft Model Antitumor Assays
- Cell Proliferation/drug effects
- Drug Discovery
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Male
Collapse
Affiliation(s)
- Jianxia Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yi Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Mengying Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Peipei Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Hao Yan
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Dalong Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Jiahao Zheng
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Bin Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Chaoyue Chen
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Di Cao
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Xiaohui Zheng
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Xiaokun Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Kun Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Zhiguo Liu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| |
Collapse
|
19
|
Chen X, Jiang R, Huang X, Chen L, Hu X, Wei Y. Long Noncoding RNA NKX2-1-AS1 Accelerates Non-Small Cell Lung Cancer Progression through the miR-589-5p/NME1 Axis. Cell Biochem Biophys 2025; 83:415-427. [PMID: 39117986 DOI: 10.1007/s12013-024-01472-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the most common malignancy worldwide, with a high death rate. Long noncoding RNA (LncRNA) NKX2-1 antisense RNA 1 (NKX2-1-AS1) has been reported to be an oncogene in lung tumorigenesis. However, the precise mechanism of NKX2-1-AS1 underlying NSCLC progression is blurry. The intention of our research was to probe the potential mechanism of NKX2-1-AS1 underlying NSCLC. NKX2-1-AS1 expression and relevant downstream gene expression were measured using RT-qPCR. Cell proliferation and apoptosis were determined by MTT assay, EdU assay along with flow cytometry analysis. Cell migratory and invasive abilities were inspected by transwell assay. Western blot and immunofluorescence staining were utilized to assess the levels of epithelial-mesenchymal transition (EMT)-related proteins. RNA pull-down together with luciferase reporter assays were performed to verify the interaction between NKX2-1-AS1 and its downstream RNAs. Xenograft tumor-bearing mouse models were built to analyze tumor growth in vivo. The results suggested that NKX2-1-AS1 was upregulated in NSCLC patient tissues and cell lines. NKX2-1-AS1 deficiency suppressed cell proliferation, migration, invasion and EMT while elevated apoptosis. NKX2-1-AS1 bound to miR-589-5p, and NME/NM23 nucleoside diphosphate kinase 1 (NME1) was targeted by miR-589-5p in NSCLC cells. Additionally, NKX2-1-AS1 accelerated the progression of NSCLC by regulating miR-589-5p/NME1 axis. NKX2-1-AS1 knockdown repressed tumor growth in vivo. In conclusion, NKX2-1-AS1 accelerated the NSCLC progression through interacting with miR-589-5p for NME1 upregulation, which may provide clues for NSCLC targeting therapy.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Ruilai Jiang
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Xiaocheng Huang
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Ling Chen
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Xiaogang Hu
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China
| | - Yanbin Wei
- Department of Respiratory and Critical Care Medicine, The Second Peoples' Hospital of Lishui, Lishui, China.
| |
Collapse
|
20
|
Jiang X, Chen J, Ding S, Yin J, Gu J, Fang X. The expression of canopy FGF signaling regulator 2 serves as a diagnostic and prognostic indicator for NSCLC. Clin Biochem 2025; 136:110895. [PMID: 39938731 DOI: 10.1016/j.clinbiochem.2025.110895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. The identification of new biomarkers is crucial for enhancing early detection and treatment outcomes. This study explores the role of Canopy FGF Signaling Regulator 2 (CNPY2) in NSCLC progression and its potential as a diagnostic and prognostic biomarker. METHODS CNPY2 expression was analyzed in 228 NSCLC tumor samples and adjacent normal tissues using quantitative RT-PCR and ELISA. Serum CNPY2 levels were also measured in 160 healthy controls and NSCLC patients. The relationship between CNPY2 expression and clinicopathological features, including epithelial-mesenchymal transition (EMT) markers, was assessed. Receiver operator curve analysis was used to evaluate the diagnostic potential of serum CNPY2, while Kaplan-Meier survival analysis assessed its prognostic significance. RESULTS CNPY2 levels were significantly elevated in NSCLC tissues compared to adjacent normal tissues. Higher CNPY2 expression was associated with larger tumor size, advanced T stage, and higher N stage. Furthermore, CNPY2 expression was positively correlated with Vimentin and N-cadherin, and negatively correlated with E-cadherin. Elevated serum CNPY2 levels in NSCLC patients demonstrated moderate diagnostic accuracy, with an area under the curve of 0.78. High CNPY2 expression was also linked to reduced overall survival (p = 0.001). CONCLUSIONS CNPY2 is markedly overexpressed in NSCLC and is associated with increased tumor aggressiveness and EMT. Serum CNPY2 shows promise as a non-invasive biomarker for NSCLC diagnosis, and elevated expression is correlated with a poorer prognosis. Thus, CNPY2 may serve as both a valuable biomarker and a potential therapeutic target in NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/diagnosis
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/blood
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Female
- Male
- Middle Aged
- Lung Neoplasms/diagnosis
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Lung Neoplasms/blood
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Prognosis
- Epithelial-Mesenchymal Transition
- Aged
- Gene Expression Regulation, Neoplastic
- Adaptor Proteins, Signal Transducing/blood
- Adaptor Proteins, Signal Transducing/genetics
- Adult
- ROC Curve
Collapse
Affiliation(s)
- Xiao Jiang
- Department of Ultrasonography, Shanghai Yangpu District Shidong Hospital, No. 999 Shiguang Road, Shanghai 200438 China
| | - Jun Chen
- Department of Ultrasonography, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi 214023 China
| | - Shujun Ding
- Department of Ultrasonography, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi 214023 China
| | - Jun Yin
- Department of Ultrasonography, Shanghai Yangpu District Shidong Hospital, No. 999 Shiguang Road, Shanghai 200438 China
| | - Jiying Gu
- Department of Ultrasonography, Shanghai Yangpu District Shidong Hospital, No. 999 Shiguang Road, Shanghai 200438 China.
| | - Xiangming Fang
- Department of Radiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi 214023 China.
| |
Collapse
|
21
|
ZHOU DAIEN, YUAN HAOYANG, HU YIWEI, WANG CHUXU, GE SA, SHAO KOUFENG, WANG HONGYING, TIAN XIAOFENG, HU HAIBO. Loss of TNFRSF21 induces cisplatin sensitivity in lung adenocarcinoma. Oncol Res 2025; 33:653-663. [PMID: 40109864 PMCID: PMC11915077 DOI: 10.32604/or.2024.050182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/07/2024] [Indexed: 03/22/2025] Open
Abstract
Background Despite the identification of numerous therapeutic targets in lung cancer, achieving significant efficacy has been challenging. TNFRSF21 plays an important role in various cancers. We investigated the function of TNFRSF21 in lung adenocarcinoma (LUAD). Methods The prognostic value of TNFRSF21 expression in lung cancer was evaluated by the GEPIA and Kaplan-Meier Plotter databases. Lung cancer cell viability was assessed by the CCK8 assay. TNFRSF21 expression patterns in lung cancer tissues and cells were examined using RT-PCR assay. Tumor sphere growth was evaluated through tumor sphere formation assays. MtROS contents in lung cancer cells were observed through MitoSOX fluorescent assays. Result TNFRSF21 was up-regulated in LUAD patients. TNFRSF21 induction was particularly notable in LUAD, especially in cancerous cells (A549, H1299, H460, and SPC-A1), compared to BEAS-2B cells. Additionally, TNFRSF21 was increased in cisplatin (DDP)-resistant LUAD cells. Loss of TNFRSF21 significantly inhibited LUAD cell growth. It was observed that forced expression of TNFRSF21 contributed to tumor cell proliferation and DDP resistance. The production of ROS was found to participate in the inhibitory effects on lung cancer stem cells (CSCs), with decreased TNFRSF21 restraining ROS contents. Collectively, these findings reveal that the downregulation of TNFRSF21 promotes ROS contents to restrain the lung CSC-like characteristics via modulation of CD44 and CD133. Conclusions In conclusion, TNFRSF21 may act as a novel target for lung cancer chemotherapy, particularly for eradicating lung CSCs.
Collapse
Affiliation(s)
- DAIEN ZHOU
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - HAOYANG YUAN
- Medical Faculty, Kunming University of Science and Technology, Kunming, 650000, China
| | - YIWEI HU
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - CHUXU WANG
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - SA GE
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - KOUFENG SHAO
- Department of Oncology, Huai’an Chuzhou Hospital of Traditional Chinese Medicine, Zhongda Hospital Group Hospital Affiliated to Southeast University, Huai’an, 223001, China
| | - HONGYING WANG
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - XIAOFENG TIAN
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - HAIBO HU
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| |
Collapse
|
22
|
Liu J, Zhao T, Sun Z, Wang J, Chai Z, Chen G. Single-cell profiling and clinical characteristics analysis of lung squamous carcinoma. Funct Integr Genomics 2025; 25:45. [PMID: 40014154 DOI: 10.1007/s10142-025-01556-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
Lung squamous carcinoma (LUSC) is a highly heterogeneous disease. However, the tumor microenvironment (TME) landscape and clinical characteristics for LUSC have not yet been elucidated. To map the TME and clinical characteristics of LUSC, we performed single-cell RNA sequencing for 504 LUSC samples on basis of TCGA and Gene Expression Omnibus. We introduced the computational algorithms "ESTIMATE" and "CIBERSORT" to analyze immune cell infiltration and immune-checkpoint-related gene signatures in various LUSC clusters. Weighted gene co-expression network analysis was used to explore the connections between molecular characteristics and clinical traits in LUSC. A prognostic model was constructed by performing multivariate COX. Two gene clusters exhibiting disparate immune and clinical characteristics were identified. Our findings indicate that patients in cluster 2, who have a more favorable prognosis, exhibit immune characteristics such as elevated levels of immunosuppression-associated M2 macrophages, resting memory CD4 T cells, resting dendritic cells (DC), and TNFRSF4, alongside reduced infiltration of activated DC and lower expression of TNFRSF18.Whereafter, the Risk Score model was built on basis of 3-DEGs signature consisted of cystatin C (CST3), transglutaminase type 2 (TGM2), JUN, which were proved by q-PCR and immunofluorescence. Besides, high-Risk Score may be responsible for poor prognosis in LUSC patients. Our study identified that tumor-infiltrating immune cell subtypes and the Risk Score model might shed light on the heterogeneity in LUSC patients. The TME, three DEGs and Risk Score can effectively serve as biomarkers to elucidate the immune landscape and predict prognosis in LUSC patients. They may provide insights to the investigations on therapeutic strategies for LUSC.
Collapse
Affiliation(s)
- Jie Liu
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China.
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China.
| | - Tian Zhao
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Zhengliang Sun
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Jinyi Wang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Zhengjun Chai
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Guohan Chen
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China.
| |
Collapse
|
23
|
Qin W, You T, Guo T, Tian R, Cui X, Wang P. The Inter-Relationships Among the Risk Factors for Pulmonary Infection and the Diagnostic Utility of Inflammatory Markers in Patients with Non-Small Cell Lung Cancer. Infect Drug Resist 2025; 18:1111-1123. [PMID: 40027913 PMCID: PMC11871871 DOI: 10.2147/idr.s491507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
Objective To analyze independent risk factors and the distribution of pathogenic bacteria associated with pulmonary infection (PI) in patients with non-small cell lung cancer (NSCLC); and to explore the utility of peripheral blood-related inflammatory markers in the diagnosis of PI after comprehensive treatment of NSCLC. Methodology We retrospectively analyzed patients who were first diagnosed with NSCLC and received anti-tumor therapy from January 1, 2022 to December 31, 2022. There were 150 males and 50 females enrolled, and with a median age of 63 years. They were divided into PI (n=59) and non-PI (n=141) groups. The patients' characteristics, treatment histories, and peripheral blood inflammatory indicators of the two groups were compared. Results Eighty-two strains of pathogens were isolated from 59 patients with NSCLC complicated by PI, of which 35 (42.7%) were Gram-negative and 29 (35.4%) were Gram-positive bacteria, 15 (18.3%) were fungi, and 3 (3.7%) were viruses. Logistic regression analysis revealed that ≥ 4 cycles of chemotherapy, radiotherapy, and higher neutrophil count before antineoplastic treatment were independent risk factors for PI in patients with NSCLC (P<0.05). C-reactive protein (CRP) and CRP-to-albumin ratio (CAR) exhibited high area under the curve values of 0.807 and 0.791, respectively. CRP demonstrated high sensitivity (Cutoff Value:10.095, P<0.001), whereas CAR had high specificity (Cutoff Value:0.747, P<0.001) for the early diagnosis of PI in patients with NSCLC. Conclusion Gram-negative bacteria were the main pathogens causing PI in patients with NSCLC. Greater than or equal to 4 cycles of chemotherapy, radiotherapy, and elevated neutrophil count before antineoplastic treatment were independent risk factors for PI in patients with NSCLC. Elevated CRP levels and higher CAR demonstrated a certain diagnostic utility for PI in patients with NSCLC after antitumor treatment.
Collapse
Affiliation(s)
- Wenwen Qin
- Department of Respiratory Medicine, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People’s Republic of China
| | - Tiebin You
- Department of Respiratory Medicine, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People’s Republic of China
| | - Tai Guo
- Department of Respiratory Medicine, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People’s Republic of China
| | - Ruixin Tian
- Department of Respiratory Medicine, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People’s Republic of China
| | - Xiaoman Cui
- Department of Respiratory Medicine, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People’s Republic of China
| | - Ping Wang
- Department of Respiratory Medicine, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, People’s Republic of China
| |
Collapse
|
24
|
Cai P, Sun H, Jiang T, Li H, Huang D, Hao X, Wang W, Xing W, Liang G. Harnessing TAGAP to improve immunotherapy for lung squamous carcinoma treatment by targeting c-Rel in CD4+ T cells. Cancer Immunol Immunother 2025; 74:114. [PMID: 39998561 PMCID: PMC11861500 DOI: 10.1007/s00262-025-03960-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/27/2025] [Indexed: 02/27/2025]
Abstract
Revealing the immunosenescence, particularly in CD4+ T cell function in lung squamous carcinoma (LUSC) assists in devising individual treatment strategies. This study identifies differentially expressed genes (DEGs) between ROS1 mutated (ROS1MUT) and wild-type (ROS1WT) LUSC samples from the TCGA database. Using WGCNA, immune-related DEGs (IRGs) were screened. Prognostic signatures derived from IRGs were used to compare immune infiltration, chemotherapy sensitivity, and immune-phenotyping score (IPS) between high- and low-risk subgroups. Hub gene abundance in different cell clusters was analyzed via Sc-seq. TAGAP overexpression or silencing was employed to assess its impact on cytokines production and differentiation of CD4+ T cells, downstream c-Rel expression, and tumor progression. High-risk subgroups exhibited decreased infiltration of natural killer, follicular helper T, and CD8+ T cells, but increased plasma, CD4+ memory resting T, and macrophage M2 cells. These subgroups were more sensitive to Sunitinib and CTLA4 blockade. TAGAP expression was significantly reduced in LUSC. Overexpressing TAGAP enhanced CD4+ T cells to produce cytokines, promoted differentiation into Th1/Th17 cells, inhibited Treg conversion, and suppressed LUSC cell phenotype in vitro. TAGAP overexpression in CD4+ T cells also inhibited LUSC tumor growth and boosted immune infiltration in vivo. TAGAP's effects on CD4+ T cells were partly reversed by c-Rel overexpression, highlighting TAGAP's role in rejuvenating CD4+ T cells and exerting anticancer effects by inhibiting c-Rel. This study elucidates the novel therapeutic potential of targeting TAGAP to modulate CD4+ T cell activity in immunotherapy for LUSC.
Collapse
Affiliation(s)
- Peian Cai
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Haibo Sun
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Tongmeng Jiang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, College of Emergency and Trauma, Hainan Provincial Stem Cell Research Institute, Hainan Medical University, Haikou, 571199, China.
| | - Huawei Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Dejing Huang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xiaopei Hao
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Wei Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Wenqun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Guanghui Liang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
25
|
Flores Banda JS, Gangane S, Raza F, Massarelli E. Current Development of Therapeutic Vaccines in Lung Cancer. Vaccines (Basel) 2025; 13:185. [PMID: 40006732 PMCID: PMC11860707 DOI: 10.3390/vaccines13020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer vaccines have a potential to change the current landscape of immunotherapy research and development. They target and neutralize specific tumor cells by utilizing the body's own immune system which offers a promising modality in treating various cancers including lung cancer. Historically, prior vaccination approaches specifically towards lung cancer have posed several challenges but also potential with early phase I/II trials showing improved overall survival. With better understanding of the body's immune system as well as advancements in vaccine development, the use of vaccines to target lung cancer cells in both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) has shown promise but also challenges in the setting of advanced stage cancers, tumor resistance mechanisms, immune evasion, and tumor heterogeneity. The proposed solution is to enroll patients in the early stages of the disease, rather than waiting until progression occurs. Additionally, future efforts will focus on the targeted identification of specific and novel tumor neo-antigens. This review offers discussion and analysis of both completed and ongoing trials utilizing different strategies for vaccine development in relation to treating lung cancer as well as current challenges faced.
Collapse
Affiliation(s)
| | | | | | - Erminia Massarelli
- Department of Medicine, University of Texas at Tyler School of Medicine, 11937 US Hwy 271, Tyler, TX 75799, USA; (J.S.F.B.); (S.G.); (F.R.)
| |
Collapse
|
26
|
Han Y, Liu S, Zhu J, Liu P, Meng Z, Li Y, Li S, Fan F, Zhang M, Liu H. Experimental study on the inhibitory effect of Halofuginone on NSCLC. Eur J Pharmacol 2025; 988:177221. [PMID: 39710292 DOI: 10.1016/j.ejphar.2024.177221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
In recent decades, significant advancements have been achieved in non-small cell lung cancer (NSCLC) treatment. However, drug resistance, postoperative recurrence, distant metastasis, and other critical issues arise during NSCLC treatment. Natural products play a crucial role in the development of anti-tumor drugs. Halofuginone (HF) is a derivative of Febrifugine, an extract of Dichroa febrifuga Lour, a traditional Chinese medicine. Recent studies on HF have demonstrated its antitumor activity and great potential for clinical applications. However, its antitumor effects and mechanisms in NSCLC remain unknown. This study aimed to elucidate the antitumor effect of HF on NSCLC and preliminarily explore its mechanism of action. The proliferation-related assay revealed that HF could inhibit the proliferation of lung adenocarcinoma cells HCC827 and H1975. Network pharmacology of HF and NSCLC indicated that HF could induce cellular oxidative stress, and the antitumor effect was related to autophagy, apoptosis, and cell cycle arrest. Experimental analysis using flow cytometry and western blotting confirmed that HF indeed induced autophagy, enhanced apoptosis, and caused cell cycle arrest. The addition of N-acetyl-cysteamine acid inhibits the HF-induced increase in reactive oxygen species levels, inhibits autophagy and apoptosis, and alters cell cycle distribution. The HCC827 transplantation tumor animal model established that HF substantially inhibited the growth of transplanted tumors. These findings suggest that HF exerts a significant inhibitory effect on NSCLC in vivo and in vitro. The inhibitory effect of HF on NSCLC was associated with the increase of ROS in tumor cells, induction of autophagy and apoptosis, and cell cycle arrest.
Collapse
Affiliation(s)
- Yuehua Han
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Shiyao Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Juan Zhu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Peipei Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Zixuan Meng
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Yongping Li
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Shanshan Li
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Fangtian Fan
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Mengxiao Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China.
| | - Hao Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China.
| |
Collapse
|
27
|
Desharnais L, Sorin M, Rezanejad M, Liu B, Karimi E, Atallah A, Swaby AM, Yu MW, Doré S, Hartner S, Fiset B, Wei Y, Kadang B, Rayes R, Joubert P, Camilleri-Broët S, Fiset PO, Quail DF, Spicer JD, Walsh LA. Spatially mapping the tumour immune microenvironments of non-small cell lung cancer. Nat Commun 2025; 16:1345. [PMID: 39905080 PMCID: PMC11794701 DOI: 10.1038/s41467-025-56546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths. An enhanced understanding of the immune microenvironments within these tumours may foster more precise and efficient treatment, particularly for immune-targeted therapies. The spatial architectural differences between lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are relatively unexplored. Here, we applied imaging mass cytometry to a balanced cohort of LUAD and LUSC patients, matched for clinical factors such as age, sex, and smoking history, to analyze 204 histopathology images of tumours from 102 individuals with non-small cell lung cancer (NSCLC). By analyzing interactions and broader cellular networks, we interrogate the tumour microenvironment to understand how immune cells are spatially organized in clinically matched adenocarcinoma and squamous cell carcinoma subsets. This spatial analysis revealed distinct patterns of immune cell aggregation, particularly among macrophage populations, that correlated with patient prognosis differentially in adenocarcinoma and squamous cell carcinoma, suggesting potential new strategies for therapeutic intervention. Our findings underscore the importance of analyzing NSCLC histological subtypes separately when investigating the spatial immune landscape, as microenvironmental characteristics and cellular interactions differed by subtype. Recognizing these distinctions is essential for designing precision therapies tailored to each subtype's unique immune architecture, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Lysanne Desharnais
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mark Sorin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Morteza Rezanejad
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Bridget Liu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Aline Atallah
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Anikka M Swaby
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Miranda W Yu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Samuel Doré
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Saskia Hartner
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Yuhong Wei
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Baharak Kadang
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Roni Rayes
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Philippe Joubert
- Département de biologie moléculaire, biochimie médicale et de pathologie, Laval University, Québec City, QC, Canada
| | | | | | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Jonathan D Spicer
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Surgery, McGill University Health Center, Montreal, QC, Canada.
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
28
|
Baldari S, Antonini A, Di Rocco G, Toietta G. Expression pattern and prognostic significance of aldehyde dehydrogenase 2 in lung adenocarcinoma as a potential predictor of immunotherapy efficacy. CANCER INNOVATION 2025; 4:e149. [PMID: 39640071 PMCID: PMC11620833 DOI: 10.1002/cai2.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 12/07/2024]
Abstract
Background The incidence of alcohol-associated cancers is higher within Asian populations having an increased prevalence of an inactivating mutation in aldehyde dehydrogenase 2 (ALDH2), a mitochondrial enzyme required for the clearance of acetaldehyde, a cytotoxic metabolite of ethanol. The role of alcohol consumption in promoting lung cancer is controversial, and little attention has been paid to the association between alcohol drinking and pulmonary ALDH2 expression. Methods We performed a comprehensive bioinformatic analysis of multi-omics data available in public databases to elucidate the role of ALDH2 in lung adenocarcinoma (LUAD). Results Transcriptional and proteomic data indicate a substantial pulmonary expression of ALDH2, which is functional for the metabolism of alcohol diffused from the bronchial circulation. ALDH2 expression is higher in healthy lung tissue than in LUAD and inhibits cell cycle, apoptosis, and epithelial-mesenchymal transition pathways. Moreover, low ALDH2 mRNA levels predict poor prognosis and low overall survival in LUAD patients. Interestingly, ALDH2 expression correlates with immune infiltration in LUAD. Conclusions A better understanding of the role of ALDH2 in lung tumor progression and immune infiltration might support its potential use as a prognostic marker and therapeutic target for improving immunotherapeutic response.
Collapse
Affiliation(s)
- Silvia Baldari
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Annalisa Antonini
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic TargetsIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Gabriele Toietta
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| |
Collapse
|
29
|
Shi Y, Zhang H, Chen H, Guo J, Yuan R, Tian Y, Xin Q, Mu Z, Tao Y, Chu Y, Wang A, Zhang Z, Tian J, Wang H. Cetuximab-Immunoliposomes Loaded with TGF-β1 siRNA for the Targeting Therapy of NSCLC: Design, and In Vitro and In Vivo Evaluation. Int J Mol Sci 2025; 26:1196. [PMID: 39940962 PMCID: PMC11818693 DOI: 10.3390/ijms26031196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Transforming growth factor-β1 (TGF-β1) promotes the growth and metastasis of lung cancer cells. Therefore, TGF-β1 siRNA (siTGF-β1) gene therapy was introduced to inhibit the expression of TGF-β1 at the nucleic acid level to avert tumor growth and metastasis. However, the delivery of naked siRNA is typically restricted by a short half-life in vivo, difficulties in delivery in vivo, and safety issues. Using siTGF-β1 as a model drug, we established an actively targeted immunoliposome delivery system to investigate the role of siTGF-β1 in non-small-cell lung cancer (NSCLC). The results showed that the constructed immune liposomes were in a position to deliver siTGF-β1 to tumor cells, thus achieving a series of effects such as improving the poor stability and short half-life of naked siRNA. RNA interference of siTGF-β1 reduced the cell viability, growth, and migration potential of human non-small cell lung cancer cells (A549). Moreover, in an A549 tumor-bearing nude mouse model, siTGF-β1 transfection markedly reduced tumor growth and tumor volume. Inhibiting TGF-β1 diminished cancer cell viability and migration and promoted apoptosis in NSCLC, as confirmed by the findings of this study. Therefore, targeting siTGF-β1 with immunoliposomes may be a new therapeutic strategy for treating non-small-cell lung cancer.
Collapse
Affiliation(s)
- Yanan Shi
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Houqian Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Hao Chen
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Jianwei Guo
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai 264006, China;
| | - Ranran Yuan
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Yu Tian
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Quanlin Xin
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Zhen Mu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Yuping Tao
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Yongchao Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Aiping Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Zhiwen Zhang
- School of Pharmacy, Fudan University, Shanghai 200437, China;
| | - Jingwei Tian
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (H.Z.); (H.C.); (R.Y.); (Y.T.); (Q.X.); (Z.M.); (Y.T.); (Y.C.); (A.W.); (J.T.)
| |
Collapse
|
30
|
Wang Q, Sun N, Li J, Huang F, Zhang Z. Liquid-Liquid Phase Separation in the Prognosis of Lung Adenocarcinoma: An Integrated Analysis. Curr Cancer Drug Targets 2025; 25:323-334. [PMID: 39506421 DOI: 10.2174/0115680096345676241001081051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a highly lethal malignancy. Liquid- Liquid Phase Separation (LLPS) plays a crucial role in targeted therapies for lung cancer and in the progression of lung squamous cell carcinoma. However, the role of LLPS in the progression and prognosis of LUAD remains insufficiently explored. METHODS This study employed a multi-step approach to identify LLPS prognosis-related genes in LUAD. First, differential analysis, univariate Cox regression analysis, Random Survival Forest (RSF) method, and Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression analysis were utilized to identify five LLPS prognosis-related genes. Subsequently, LASSO Cox regression was performed to establish a prognostic score termed the LLPS-related prognosis score (LPRS). Comprehensive analyses were then conducted based on the LPRS, including survival analysis, clinical feature analysis, functional enrichment analysis, and tumor microenvironment assessment. The LPRS was integrated with additional clinicopathological factors to develop a prognostic nomogram for LUAD patients. Immunohistochemical validation was performed on clinical tissue samples to further validate the findings. Finally, the relationship between KRT6A, one of the identified genes, and epidermal growth factor receptor (EGFR) mutations was investigated. RESULTS The LPRS was established using five LLPS-related genes: IGF2BP1, KRT6A, LDHA, PKP2, and PLK1. Higher LPRS was closely associated with poor survival outcomes, gender, progression-free survival (PFS), and advanced TNM stage. Furthermore, LPRS emerged as an independent prognostic factor for LUAD. A nomogram integrating LPRS, TNM stage, and age demonstrated remarkable predictive accuracy for prognosis among patients with LUAD. LLPS likely influences LUAD prognosis through the activity of IGF2BP1, KRT6A, LDHA, PKP2, and PLK1. KRT6A exhibits significant upregulation in LUAD, particularly in patients with EGFR mutations. CONCLUSION This study introduces a novel LPRS model that demonstrates high accuracy in predicting the clinical prognosis of LUAD. Moreover, the findings suggest that KRT6A may play a critical role in the LLPS-mediated malignant progression of LUAD.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Respiration, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Nannan Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jianhao Li
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Fengxiang Huang
- Department of Respiration, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhao Zhang
- Department of Respiration, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
31
|
Tanzhu G, Chen L, Ning J, Xue W, Wang C, Xiao G, Yang J, Zhou R. Metastatic brain tumors: from development to cutting-edge treatment. MedComm (Beijing) 2025; 6:e70020. [PMID: 39712454 PMCID: PMC11661909 DOI: 10.1002/mco2.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 12/24/2024] Open
Abstract
Metastatic brain tumors, also called brain metastasis (BM), represent a challenging complication of advanced tumors. Tumors that commonly metastasize to the brain include lung cancer and breast cancer. In recent years, the prognosis for BM patients has improved, and significant advancements have been made in both clinical and preclinical research. This review focuses on BM originating from lung cancer and breast cancer. We briefly overview the history and epidemiology of BM, as well as the current diagnostic and treatment paradigms. Additionally, we summarize multiomics evidence on the mechanisms of tumor occurrence and development in the era of artificial intelligence and discuss the role of the tumor microenvironment. Preclinically, we introduce the establishment of BM models, detailed molecular mechanisms, and cutting-edge treatment methods. BM is primarily treated with a comprehensive approach, including local treatments such as surgery and radiotherapy. For lung cancer, targeted therapy and immunotherapy have shown efficacy, while in breast cancer, monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates are effective in BM. Multiomics approaches assist in clinical diagnosis and treatment, revealing the complex mechanisms of BM. Moreover, preclinical agents often need to cross the blood-brain barrier to achieve high intracranial concentrations, including small-molecule inhibitors, nanoparticles, and peptide drugs. Addressing BM is imperative.
Collapse
Affiliation(s)
- Guilong Tanzhu
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Liu Chen
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jiaoyang Ning
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Wenxiang Xue
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunJilinChina
| | - Ce Wang
- Department of RadiologyChina‐Japan Friendship HospitalBeijingChina
| | - Gang Xiao
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jie Yang
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Department of DermatologyXiangya HospitalCentral South UniversityChangshaChina
| | - Rongrong Zhou
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Xiangya Lung Cancer CenterXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan ProvinceChina
| |
Collapse
|
32
|
Pan H, Zhang H, Zhang Y, Chen X, Liu Z, Wu Y, Bai N, Shi Y, Zhao M, Zhu L. Genetic profile in primary tumor tissue of advanced lung adenocarcinoma patients with adrenal metastasis. Cancer Genet 2025; 290-291:36-43. [PMID: 39673828 DOI: 10.1016/j.cancergen.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/07/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
The aim of this study was to examine the genomic characteristics and explore the molecular mechanisms underlying adrenal metastases in lung adenocarcinoma. 57 patients diagnosed with lung adenocarcinoma (LUAD) and adrenal metastases (AM) were enrolled, alongside 33 controls diagnosed with non-adrenal metastases (non-AM) at the time of diagnosis. The primary lung cancer tissue sample were analyzed using next-generation sequencing. The molecular and clinical features were correlated with clinical outcomes. TP53, EGFR and KRAS were the most frequently mutated gene in both groups. EGFR mutations, especially rare variants (G724A, L747P, Q701 L, G719C, V769 L and S768I), exhibited significant enrichment in the non-AM group (P<0.001). An elevated age-related signature in the group of patients with AM, whereas the non-AM group exhibited a higher BRCA signature. Potential prognostic biomarkers such as KEAP1, LRP1B, NOTCH1 and RET mutations were detected in the non-AM group, while ALK mutations in the AM group correlated with shorter overall survival (P<0.001). KRAS mutations in the early synchronous adrenal metastases group were also associated with shorter OS (P<0.001). The analysis of 425 tumor genes in 29 patients with adrenal metastases showed significant enrichment in pathways associated with invasion and metastasis, including TNF signaling pathway and TGF-β signaling pathway. Patients without EGFR mutations in LUAD need to be more concerned about adrenal metastases. Meanwhile, patients with adrenal metastases harboring ALK or KRAS mutations have a poor prognosis and require more aggressive treatment. The TNF and TGF-β pathways might be associated with adrenal metastasis.
Collapse
Affiliation(s)
- Haiqiao Pan
- Chengde Medical University, Chengde, Hebei 067000, PR China; Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China
| | - Hongbin Zhang
- Department of Oncology, Hebei Chest Hospital, Shijiazhuang, Hebei 050000, PR China
| | - Yongqian Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China
| | - Xiaojing Chen
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China; Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Zhai Liu
- Department of Radiotherapy, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Yajing Wu
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Na Bai
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, Jiangsu 210000, PR China
| | - Yan Shi
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, Jiangsu 210000, PR China
| | - Min Zhao
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China.
| | - Lingling Zhu
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China.
| |
Collapse
|
33
|
Luan X, Peng X, Hui G, Wei Z. TFAP2A Promotes Cell Progression and Suppresses Ferroptosis in Lung Adenocarcinoma via Activating Transcription of CST1. J Biochem Mol Toxicol 2025; 39:e70087. [PMID: 39692484 DOI: 10.1002/jbt.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/28/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
Lung adenocarcinoma (LUAD) is a common type of lung cancer with complicated pathological mechanism. Transcription Factor AP-2 Alpha (TFAP2A) and Cysteine protease inhibitor 1 (CST1) are upregulated genes in LUAD samples, accordingly, we focused on clarifying the role of TFAP2A/CST1 axis in LUAD. Expression analysis was performed using real-time quantitative polymerase chain reaction and western blot. Cellular behaviors were detected by colony formation assay, EdU assay, wound healing assay and flow cytometry. Ferroptosis was assessed by oxidative indicators, Fe2+ level and related proteins. TFAP2A and CST1 interaction was analyzed via ChIP assay and dual-luciferase reporter assay. TFAP2A function in vivo was evaluated by xenograft tumor assay. CST1 was overexpressed in LUAD samples and cells. Downregulation of CST1 inhibited proliferation, migration but it promoted apoptosis and ferroptosis of LUAD cells. TFAP2A interacted with the promoter of CST1 to up-regulate CST1 expression. TFAP2A regulated the malignant behaviors and ferroptosis of LUAD cells by targeting CST1. TFAP2A affected LUAD tumor growth via mediating CST1. All these data proved that TFAP2A/CST1 axis contributed to proliferation, migration while it suppressed apoptosis and ferroptosis in LUAD.
Collapse
Affiliation(s)
- Xinyu Luan
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xuxing Peng
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Gang Hui
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zichun Wei
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
34
|
Li G, Li Q, Tong Y, Zeng J, Dang T, Yang N, Zhou Y, Ma L, Ge Q, Zhao Z. The anticancer mechanisms of Toxoplasma gondii rhoptry protein 16 on lung adenocarcinoma cells. Cancer Biol Ther 2024; 25:2392902. [PMID: 39174877 PMCID: PMC11346528 DOI: 10.1080/15384047.2024.2392902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 04/29/2024] [Accepted: 08/11/2024] [Indexed: 08/24/2024] Open
Abstract
Lung adenocarcinoma is the most prevalent subtype of lung cancer, which is the leading cause of cancer-related mortality worldwide. Toxoplasma gondii (T.gondii) Rhoptry protein 16 (ROP16) has been shown to quickly enter the nucleus, and through activate host cell signaling pathways by phosphorylation STAT3 and may affect the survival of tumor cells. This study constructed recombinant lentiviral expression vector of T. gondii ROP16 I/II/III and stably transfected them into A549 cells, and the effects of ROP16 on cell proliferation, cell cycle, apoptosis, invasion, and migration of A549 cells were explored by utilizing CCK-8, flow cytometry, qPCR, Western blotting, TUNEL, Transwell assay, and cell scratch assay, and these effects were confirmed in the primary human lung adenocarcinoma cells from postoperative cancer tissues of patients. The type I and III ROP16 activate STAT3 and inhibited A549 cell proliferation, regulated the expression of p21, CDK6, CyclinD1, and induced cell cycle arrest at the G1 phase. ROP16 also regulated the Bax, Bcl-2, p53, cleaved-Caspase3, and Caspase9, inducing cell apoptosis, and reduced the invasion and migration of A549 cells, while type II ROP16 protein had no such effect. Furthermore, in the regulation of ROP16 on primary lung adenocarcinoma cells, type I and III ROP16 showed the same anticancer potential. These findings confirmed the anti-lung adenocarcinoma effect of type I and III ROP16, offering fresh perspectives on the possible application of ROP16 as a target with adjuvant therapy for lung adenocarcinoma and propelling the field of precision therapy research toward parasite treatment of tumors.
Collapse
Affiliation(s)
- Guangqi Li
- Medical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Qinhui Li
- College of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Yongqing Tong
- Department of Clinical laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jin Zeng
- College of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Tiantian Dang
- Medical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ningai Yang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yuning Zhou
- The First Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Lei Ma
- College of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Qirui Ge
- The First Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Zhijun Zhao
- Medical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
35
|
Liu L, Zhang Q, Jin S, Xie L. Prognostic value of lymph node metrics in lung squamous cell carcinoma: an analysis of the SEER database. World J Surg Oncol 2024; 22:351. [PMID: 39731070 DOI: 10.1186/s12957-024-03639-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024] Open
Abstract
INTRODUCTION Although the Tumor-Node-Metastasis (TNM) staging system is widely used for staging lung squamous cell carcinoma (LSCC), the TNM system primarily emphasizes tumor size and metastasis, without adequately considering lymph node involvement. Consequently, incorporating lymph node metastasis as an additional prognostic factor is essential for predicting outcomes in LSCC patients. METHODS This retrospective study included patients diagnosed with LSCC between 2004 and 2018 and was based on data from the Surveillance, Epidemiology, and End Results (SEER) database of the National Cancer Institute. The primary endpoint of the study was cancer-specific survival (CSS), and demographic characteristics, tumor characteristics, and treatment regimens were incorporated into the predictive model. The study focused on the value of indicators related to pathological lymph node testing, including the lymph node ratio (LNR), regional node positivity (RNP), and lymph node examination count (RNE), in the prediction of cancer-specific survival in LSCC. A prognostic model was established using a multivariate Cox regression model, and the model was evaluated using the C index, Kaplan-Meier, the Akaike information criterion (AIC), decision curve analysis (DCA), continuous net reclassification improvement (NRI), and integrated discrimination improvement (IDI), and the predictive efficacy of different models was compared. RESULTS A total of 14,200 LSCC patients (2004-2018) were divided into training and validation cohorts. The 10-year CSS rate was approximately 50%, with no significant survival differences between cohorts (p = 0.8). The prognostic analysis revealed that models incorporating LNR, RNP, and RNE demonstrated superior performance over the TNM model. The LNR and RNP models demonstrated better model fit, discrimination, and reclassification, with AUC values of 0.695 (training) and 0.665 (validation). The RNP and LNR models showed similar predictive performance, significantly outperforming the TNM and RNE models. Calibration curves and decision curve analysis confirmed the clinical utility and net benefit of the LNR and RNP models in predicting long-term CSS for LSCC patients, highlighting their value in clinical decision-making. CONCLUSION This study confirms that RNP status is an independent prognostic factor for CSS in LSCC, with predictive efficacy comparable to LNR, with both models enhancing survival prediction beyond TNM staging.
Collapse
Affiliation(s)
- Lei Liu
- School of Biology & Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, 561113, China
| | - Qiao Zhang
- Medical Department, The Second People's Hospital of Guiyang(Jinyang Hospital), Guiyang, 550081, China
| | - Shuai Jin
- School of Biology & Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, 561113, China.
| | - Lang Xie
- Department of Hospital Infection Management and Preventive Health Care, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, 551799, China.
| |
Collapse
|
36
|
Pan H, Liao L, Xu S, Xu Y, Chai W, Liu X, Li J, Cao Y, Sun L, Liu Q, Yan M. RBIS regulates ribosome biogenesis to affect progression in lung adenocarcinoma. J Transl Med 2024; 22:1147. [PMID: 39722009 DOI: 10.1186/s12967-024-05886-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Increased ribosome biogenesis is required for tumor growth. In this study, we investigated the function and underlying molecular mechanism of ribosome biogenesis factor (RBIS) in the progression of non-small cell lung cancer (NSCLC). METHODS In our study, we conducted a comprehensive analysis to identify key genes implicated in ribosome biogenesis by leveraging a Gene Set Enrichment Analysis (GSEA) dataset. Subsequently, we performed a comparative analysis of gene expression profiles by utilizing data from the Gene Expression Omnibus (GEO) datasets to ascertain differentially expressed genes (DEGs) between cancerous and adjacent non-cancerous tissues. Through the intersection of gene sets derived from GSEA and GEO, we identified a cohort of ribosome-associated genes that might exert a substantial influence on the progression of lung adenocarcinoma. Following an extensive literature review, we have identified the RBIS gene as an interesting candidate for further investigation. To elucidate the in vitro functional role of RBIS, several assays was employed, including the Transwell migration and invasion assay, wound healing assay, Cell Counting Kit-8 (CCK-8) proliferation assay, and colony formation assay. Subcutaneous and tail vein injection-based lung metastasis xenograft tumor models were used in evaluating the tumorigenic potential, growth, and metastatic spread of lung cancer cells. Flow cytometry analysis was employed to investigate cell cycle distribution and apoptotic rates. Additionally, real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was utilized to quantify the mRNA expression levels of genes. To comprehensively assess the translational efficiency of nascent proteins, we employed polysome profiling analysis to provide insights into the cellular translational landscape. Furthermore, we quantified global protein synthesis using a fluorescence-based assay to measure protein synthesis rates. The immunofluorescence technology was utilized to study the subcellular reorganization of the nucleolus. We conducted co-immunoprecipitation (Co-IP) assays followed by Western blot analysis to identify potential proteins interacted with RBIS. The half maximal inhibitory concentration (IC50) was used for evaluating the chemosensitivity of lung cancer cells to gemcitabine. Additionally, the colony formation assay was employed to assess the survival and proliferative capacity post-treatment of gemcitabine. RESULTS The database analysis showed that RBIS was upregulated in lung adenocarcinoma, and its high expression was associated with poor prognosis; Knockdown of RBIS significantly inhibited NSCLC cell migration, invasion and proliferation in vitro and xenograft tumor growth and metastasis in vivo. Additionally, knockdown of RBIS led to G0/G1 phase arrest and significantly increased apoptosis in lung adenocarcinoma cells. Mechanistically, downregulation of RBIS significantly decreased the expression of 47S ribosomal RNA (rRNA), a component associated with ribosome assembly. Polysome profiling analysis indicated that RBIS knockdown affected protein translation efficiency, and global protein synthesis assay further verified that RBIS knockdown inhibited synthesis of newborn proteins. Additionally, the ribosomal biogenesis-targeting drugs CX-5461 and the loss of RBIS exhibited synergistic effects in inhibiting cell cycle progression and inducing apoptosis. Furthermore, the ribosomal maturation factor GNL2 was identified as the key downstream regulator of RBIS in ribosome biogenesis. Notably, knockdown of RBIS substantially increased the sensitivity of lung adenocarcinoma cells to the chemotherapeutic drug gemcitabine, highlighting its l role in chemotherapy. CONCLUSIONS Collectively, these studies suggested the close involvement of RBIS in the progression of lung adenocarcinoma, providing new insights for targeted therapeutic interventions involving ribosomes.
Collapse
Affiliation(s)
- Hongyu Pan
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Li Liao
- Department of Oncology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Siwei Xu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yujian Xu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenjun Chai
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoli Liu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jing Li
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yue Cao
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lei Sun
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Liu
- Department of Respiratory Medicine, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, 1665 Kong Jiang Road, Shanghai, 200092, China.
| | - Mingxia Yan
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
37
|
Sun T, Yang Z. TFAP2C-mediated transcriptional activation of STEAP3 promotes lung squamous cell carcinoma progression by regulating the β-catenin pathway. Biol Direct 2024; 19:135. [PMID: 39716275 DOI: 10.1186/s13062-024-00584-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
Abstract
Six-transmembrane epithelial antigen of prostate 3 (STEAP3) is associated with the progression of several human malignancies. However, its role in lung squamous cell carcinoma (LUSC) remains unclear. We measured STEAP3 expression in LUSC cell lines and tissues. LUSC cells with stable STEAP3 overexpression and knockdown were obtained through G418 selection. Multiple assays were used to evaluate the malignant phenotypes of LUSC cells and the activation of the β-catenin signaling. The potential transcriptional regulatory factors of STEAP3 were predicted using the JASPAR database, and the correlation between transcription factor AP-2 gamma (TFAP2C) and STEAP3 was analyzed through the GEPIA database. The study evaluated the regulatory relationship between a potential transcription factor and STEAP3 through ChIP and luciferase reporter assays. Additionally, rescue assays were utilized to ascertain whether TFAP2C serves as the upstream regulatory factor of STEAP3, contributing to LUSC progression. Finally, tumor growth and metastasis were evaluated in vivo. STEAP3 expression was notably higher in LUSC, and its overexpression was linked to a poor prognosis. Moreover, STEAP3 overexpression activated the β-catenin pathway, thereby accelerating cell proliferation and metastasis. Conversely, STEAP3 knockdown had an anti-tumor effect in LUSC. Additionally, TFAP2C bound directly to the STEAP3 promoter and positively regulate its expression in LUSC. The anti-tumor effects of TFAP2C knockdown were partially reversed by STEAP3 overexpression. The study indicates that the TFAP2C/STEAP3 axis may be a therapeutic target for LUSC treatment. This enhances our understanding of lung carcinogenesis.
Collapse
Affiliation(s)
- Tong Sun
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, Liaoning, P. R. China
| | - Zhiguang Yang
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, Liaoning, P. R. China.
| |
Collapse
|
38
|
Zhao X, Wang Y, Xue M, Ding Y, Zhang H, Wang K, Ren J, Li X, Xu M, Lv J, Wang Z, Sun D. Preoperative assessment of tertiary lymphoid structures in stage I lung adenocarcinoma using CT radiomics: a multicenter retrospective cohort study. Cancer Imaging 2024; 24:167. [PMID: 39696659 DOI: 10.1186/s40644-024-00813-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE To develop a multimodal predictive model, Radiomics Integrated TLSs System (RAITS), based on preoperative CT radiomic features for the identification of TLSs in stage I lung adenocarcinoma patients and to evaluate its potential in prognosis stratification and guiding personalized treatment. METHODS The most recent preoperative chest CT thin-slice scans and postoperative hematoxylin and eosin-stained pathology sections of patients diagnosed with stage I LUAD were retrospectively collected. Tumor segmentation was achieved using an automatic virtual adversarial training segmentation algorithm based on a three-dimensional U-shape convolutional neural network (3D U-Net). Radiomic features were extracted from the tumor and peritumoral areas, with extensions of 2 mm, 4 mm, 6 mm, and 8 mm, respectively, and deep learning image features were extracted through a convolutional neural network. Subsequently, the RAITS was constructed. The performance of RAITS was then evaluated in both the train and validation cohorts. RESULTS RAITS demonstrated superior AUC, sensitivity, and specificity in both the training and external validation cohorts, outperforming traditional unimodal models. In the validation cohort, RAITS achieved an AUC of 0.78 (95% CI, 0.69-0.88) and showed higher net benefits across most threshold ranges. RAITS exhibited strong discriminative ability in risk stratification, with p < 0.01 in the training cohort and p = 0.02 in the validation cohort, consistent with the actual predictive performance of TLSs, where TLS-positive patients had significantly higher recurrence-free survival (RFS) compared to TLS-negative patients (p = 0.04 in the training cohort, p = 0.02 in the validation cohort). CONCLUSION As a multimodal predictive model based on preoperative CT radiomic features, RAITS demonstrated excellent performance in identifying TLSs in stage I LUAD and holds potential value in clinical decision-making.
Collapse
Affiliation(s)
| | - Yuhang Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Mengli Xue
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Yun Ding
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Han Zhang
- Chest hospital, Tianjin University, Tianjin, China
| | - Kai Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Jie Ren
- Department of Thoracic Surgery, Tianjin Jinnan Hospital, Tianjin, China
| | - Xin Li
- Chest hospital, Tianjin University, Tianjin, China
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Meilin Xu
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Jun Lv
- Department of Imaging, Tianjin Chest Hospital, Tianjin, China
| | - Zixiao Wang
- Department of Thoracic Surgery, Qinhuangdao First Hospital, Hebei Province, China
| | - Daqiang Sun
- Chest hospital, Tianjin University, Tianjin, China.
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China.
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
39
|
Sun Z, Ding C, Wang Y, Lu T, Song W. Plasma-Activated Medium Inhibited the Proliferation and Migration of Non-Small Cell Lung Cancer A549 Cells in 3D Culture. Int J Mol Sci 2024; 25:13262. [PMID: 39769029 PMCID: PMC11676436 DOI: 10.3390/ijms252413262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/02/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Lung cancer is the most common type of malignant tumor worldwide. Plasma-activated medium (PAM) is an innovative cancer treatment method that has received considerable scientific attention. The objective of this study is to evaluate the effects of PAM on the anti-tumor characteristics of non-small cell lung cancer (NSCLC) cells in two-dimensional (2D) and three-dimensional (3D) cultures. The effects of PAM treatment on the proliferative and migratory capabilities of A549 cells in 2D and 3D cultures were assessed using MTT, migration, invasion assays, and cell cycle, respectively. The study also investigated the impact of PAM treatment on the changes in the content of intracellular and extracellular reactive species and analyzed protein expression using the Western Blot method. PAM treatment inhibited the viability, migration, and invasion abilities of A549 cells in both 2D and 3D cultures, suppressed the epithelial-mesenchymal transition (EMT) process, and downregulated the expression of the RAS/ERK signaling pathway, which effectively inhibited tumor spheroid formation. Additionally, the effect of PAM on A549 cells was mediated through ROS-induced oxidative reactions, and PAM treatment exhibited greater cytotoxicity in 2D culture compared to 3D culture. As compared to 2D, the 3D cell culture model provides a viable in vitro cell model for studying the mechanisms of PAM treatment in lung cancer. PAM represents an effective new treatment for NSCLC.
Collapse
Affiliation(s)
- Zhidan Sun
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (Z.S.); (C.D.); (Y.W.)
- College of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Chenglong Ding
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (Z.S.); (C.D.); (Y.W.)
- College of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Yuhan Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (Z.S.); (C.D.); (Y.W.)
- College of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Tingting Lu
- Key Laboratory for the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wencheng Song
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (Z.S.); (C.D.); (Y.W.)
- College of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
- Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences, Soochow University, Suzhou 215123, China
| |
Collapse
|
40
|
Nu er lan STE, Yu B, Yang Y, Shen Y, Xu B, Zhan Y, Liu C. Discover Mutational Differences Between Lung Adenocarcinoma and Lung Squamous Cell Carcinoma and Search for More Effective Biomarkers for Immunotherapy. Cancer Manag Res 2024; 16:1759-1773. [PMID: 39678041 PMCID: PMC11645897 DOI: 10.2147/cmar.s491661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Lung cancer is a severe malignant tumor. This study aims to more comprehensively characterize lung cancer patients and identify combination markers for immunotherapy. Patients and Methods We gathered data from 166 lung cancer patients at the Cancer Hospital Affiliated with Xinjiang Medical University. The collected samples underwent NGS sequencing using a panel of 616 genes associated with cancer. Subsequently, data analysis was conducted to identify markers that are more suitable for lung cancer immunotherapy. Results In this study, the most common variant genes in LUAD were TP53, EGFR, MST1, KMT2C, RBM10, LRP1B. Meanwhile, the highest mutation frequency genes in LUSC samples were TP53, KMT2D, LRP1B, FAT1, MST1, KMT2C. Mutation frequencies, tumor mutation burden (TMB), PD-L1 expression, and mutant-allele tumor heterogeneity (MATH) values differed between LUAD and LUSC, with LUSC exhibiting higher values than LUAD. Irrespective of LUAD or LUSC, patients with TMB≥10 demonstrated better immunotherapy efficacy compared to patients with TMB<10. Similarly, when PD-L1≥50%, whether in LUAD or LUSC, the immunotherapy effect was superior to that of patients with PD-L1<50%. Combining TMB≥10 and PD-L1≥50% as immunotherapy markers, in both LUAD and LUSC, resulted in a very favorable immunotherapy effect, with the overall response rate (ORR) reaching 100%. Conclusion We observed distinct mutation patterns and clinical factors between LUAD and LUSC, and noted that patients with TMB≥10 and PD-L1≥50% exhibited enhanced immunotherapy effects. Combining TMB≥10 and PD-L1≥50% proved to be a more effective predictor of immunotherapy efficacy.
Collapse
Affiliation(s)
- Sai te er Nu er lan
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Bo Yu
- Beijing USCI Medical Laboratory, Beijing, 100195, People’s Republic of China
| | - Yan Yang
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Yanli Shen
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Bing Xu
- Beijing USCI Medical Laboratory, Beijing, 100195, People’s Republic of China
| | - Yiyi Zhan
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Chunling Liu
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| |
Collapse
|
41
|
Yang N, Yu T, Zheng B, Sun W, Li Y, Zhang W, Chen Y, Yuan L, Wang XJ, Wang J, Yang F. POSTN promotes the progression of NSCLC via regulating TNFAIP6 expression. Biochem Biophys Res Commun 2024; 736:150891. [PMID: 39471683 DOI: 10.1016/j.bbrc.2024.150891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024]
Abstract
Aberrant upregulation of Periostin (POSTN) expression has been implicated in various disease-related pathological cascades, notably inflammatory responses, fibrotic processes and tumor progression, including non-small cell lung cancer (NSCLC). The present study aimed to elucidate the functional role and underlying mechanisms of POSTN in NSCLC. Immunohistochemical and Western blot analysis consistently revealed elevated POSTN levels in NSCLC tissues and cell lines. POSTN expression negatively correlated with patient prognosis. Functional experiments utilizing POSTN-targeting siRNAs demonstrated a significant suppression of NSCLC cell proliferation, epithelial-to-mesenchymal transition (EMT), migration and invasion, whereas POSTN overexpression via plasmid transfection enhanced these oncogenic properties. Mechanistically, RNA sequencing analysis and subsequent validation studies revealed that POSTN positively modulates the transcriptional expression of tumor necrosis factor alpha-induced protein 6 (TNFAIP6) in NSCLC. Notably, a positive correlation was observed between POSTN and TNFAIP6 expression levels, and their overexpression positively correlated with NSCLC progression. Furthermore, TNFAIP6 overexpression rescued the inhibitory effects of POSTN knockdown on NSCLC malignant phenotypes. Collectively, our findings indicate that POSTN promotes NSCLC malignancy through TNFAIP6 upregulation, positioning POSTN as a promising biomarker and potential therapeutic target for NSCLC prognosis and treatment strategies in clinical settings.
Collapse
Affiliation(s)
- Nani Yang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, PR China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Tianqing Yu
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, PR China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Beiyao Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Wentao Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Yue Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Wei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Yuanyuan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Li Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Xue Jun Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China.
| | - Junbin Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, PR China.
| | - Fen Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China.
| |
Collapse
|
42
|
Wang J, He X, Jia Z, Yan A, Xiao K, Liu S, Hou M, Long Y, Ding X. Shenqi Fuzheng injection restores the sensitivity to gefitinib in non-small cell lung cancer by inhibiting the IL-22/STAT3/AKT pathway. PHARMACEUTICAL BIOLOGY 2024; 62:33-41. [PMID: 38100532 PMCID: PMC10732196 DOI: 10.1080/13880209.2023.2292266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
CONTEXT Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. Gefitinib is a first-line treatment for NSCLC. However, its effectiveness is hindered by the development of drug resistance. At present, Shenqi Fuzheng injection (SFI) is widely accepted as an adjuvant therapy in NSCLC. OBJECTIVE This study investigates the molecular mechanism of SFI when combined with gefitinib in regulating cell progression among EGFR-TKI-resistant NSCLC. MATERIALS AND METHODS We established gefitinib-resistant PC9-GR cells by exposing gefitinib escalation from 10 nM with the indicated concentrations of SFI in PC9 cells (1, 4, and 8 mg/mL). Quantitative real-time polymerase chain reaction was performed to assess gene expression. PC9/GR and H1975 cells were treated with 50 ng/mL of interleukin (IL)-22 alone or in combination with 10 mg/mL of SFI. STAT3, p-STAT3, AKT, and p-AKT expression were evaluated using Western blot. The effects on cell proliferation, clonogenicity, and apoptosis in NSCLC cells were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation and flow cytometry assays. RESULTS SFI treatment alleviated the development of gefitinib resistance in NSCLC. PC9/GR and H1975 cells treated with SFI significantly exhibited a reduction in IL-22 protein and mRNA overexpression levels. SFI effectively counteracted the activation of the STAT3/AKT signaling pathway induced by adding exogenous IL-22 to PC9/GR and H1975 cells. Moreover, IL-22 combined with gefitinib markedly increased cell viability while reducing apoptosis. In contrast, combining SFI with gefitinib and the concurrent treatment of SFI with gefitinib and IL-22 demonstrated the opposite effect. DISCUSSION AND CONCLUSION SFI can be a valuable therapeutic option to address gefitinib resistance in NSCLC by suppressing the IL-22/STAT3/AKT pathway.
Collapse
Affiliation(s)
- Jiali Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xianhai He
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhirong Jia
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Aiwen Yan
- Jiangsu Food & Pharmaceutical Science College, Jiangsu Food Science College, Huanan, China
| | - Kang Xiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shuo Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengjun Hou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yaling Long
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuansheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Precision Medicine Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
43
|
Zhang A, Gao Q, Tian C, Chen W, Pan C, Wang L, Huang J, Zhang J. Liquid Biopsy in Lung Cancer: Nano-Flow Cytometry Detection of Non-Small Cell Lung Cancer in Blood. J Transl Med 2024; 104:102151. [PMID: 39419350 DOI: 10.1016/j.labinv.2024.102151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of global mortality, with current screening and diagnostic methods often lacking in sensitivity and specificity. In our endeavor to develop precise, objective, and easily accessible diagnostic biomarkers for NSCLC, this study aimed to leverage rapidly evolving liquid biopsy techniques in the field of pathology to differentiate NSCLC patients from healthy controls by isolating peripheral blood samples and enriching extracellular vesicles (EVs) containing lung-derived proteins (thyroid transcription factor-1 [TTF-1] and surfactant protein B [SFTPB]), along with the cancer-associated protein CD151+ EVs. Additionally, for practical applications, we established a nano-flow cytometry assay to detect plasma EVs readily. NSCLC patients demonstrated significantly reduced counts of TTF-1+ EVs and CD151+ EVs in plasma compared with healthy controls (P < .0001), whereas SFTPB+ EVs showed no significant difference (P > .05). Integrated analysis of TTF-1+, CD151+, and SFTPB+ EVs yielded an area under the curve values of 0.913 and 0.854 in the discovery and validation cohorts, respectively. Thus, although further validation is essential, the newly developed technologies are of great significance for the robust detection of NSCLC biomarkers.
Collapse
Affiliation(s)
- Andong Zhang
- Department of General Medicine, The Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Qiqi Gao
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chen Tian
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wentao Chen
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Catherine Pan
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ling Wang
- Department of General Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Department of General Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Jie Huang
- Department of General Medicine, The Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Jing Zhang
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; National Health and Human Brain Tissue Resource Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
44
|
Chen S, Cheng S, Cai J, Liu Z, Li H, Wang P, Li Y, Yang F, Chen K, Qiu M. The current therapeutic cancer vaccines landscape in non-small cell lung cancer. Int J Cancer 2024; 155:1909-1927. [PMID: 39109825 DOI: 10.1002/ijc.35088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 10/04/2024]
Abstract
Currently, conventional immunotherapies for the treatment of non-small cell lung cancer (NSCLC) have low response rates and benefit only a minority of patients, particularly those with advanced disease, so novel therapeutic strategies are urgent deeded. Therapeutic cancer vaccines, a form of active immunotherapy, harness potential to activate the adaptive immune system against tumor cells via antigen cross-presentation. Cancer vaccines can establish enduring immune memory and guard against recurrences. Vaccine-induced tumor cell death prompts antigen epitope spreading, activating functional T cells and thereby sustaining a cancer-immunity cycle. The success of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rendered cancer vaccines a promising avenue, especially when combined with immunotherapy or chemoradiotherapy for NSCLC. This review delves into the intricate antitumor immune mechanisms underlying therapeutic cancer vaccines, enumerates the tumor antigen spectrum of NSCLC, discusses different cancer vaccines progress and summarizes relevant clinical trials. Additionally, we analyze the combination strategies, current limitations, and future prospects of cancer vaccines in NSCLC treatment, aiming to offer fresh insights for their clinical application in managing NSCLC. Overall, cancer vaccines offer promising potential for NSCLC treatment, particularly combining with chemoradiotherapy or immunotherapy could further improve survival in advanced patients. Exploring inhaled vaccines or prophylactic vaccines represents a crucial research avenue.
Collapse
Affiliation(s)
- Shaoyi Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Sida Cheng
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Jingsheng Cai
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Zheng Liu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Haoran Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Kezhong Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Mantang Qiu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
45
|
Shen Y, Yang Y, Zhao Y, Nuerlan S, Zhan Y, Liu C. YY1/circCTNNB1/miR-186-5p/YY1 positive loop aggravates lung cancer progression through the Wnt pathway. Epigenetics 2024; 19:2369006. [PMID: 38913848 PMCID: PMC11197906 DOI: 10.1080/15592294.2024.2369006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/12/2024] [Indexed: 06/26/2024] Open
Abstract
Lung cancer is one familiar cancer that threatens the lives of humans. circCTNNB1 has been disclosed to have regulatory functions in some diseases. However, the functions and related regulatory mechanisms of circCTNNB1 in lung cancer remain largely indistinct. The mRNA and protein expression levels were examined through real-time polymerase chain reaction (RT-qPCR) and western blot. The cell proliferation was tested through CCK-8 assay. The cell migration and invasion were confirmed through Transwell assays. The cell senescence was evaluated through SA-β-gal assay. The binding ability between miR-186-5p and circCTNNB1 (or YY1) was verified through luciferase reporter and RIP assays. In this study, the higher expression of circCTNNB1 was discovered in lung cancer tissues and cell lines and resulted in poor prognosis. In addition, circCTNNB1 facilitated lung cancer cell proliferation, migration, invasion, and suppressed cell senescence. Knockdown of circCTNNB1 retarded the Wnt pathway. Mechanism-related experiments revealed that circCTNNB1 combined with miR-186-5p to target YY1. Through rescue assays, YY1 overexpression could rescue decreased cell proliferation, migration, invasion, increased cell senescence, and retarded Wnt pathway mediated by circCTNNB1 suppression. Furthermore, YY1 acts as a transcription factor that can transcriptionally activate circCTNNB1 to form YY1/circCTNNB1/miR-186-5p/YY1 positive loop. Through in vivo assays, circCTNNB1 accelerated tumour growth in vivo. All findings revealed that a positive loop YY1/circCTNNB1/miR-186-5p/YY1 aggravated lung cancer progression by modulating the Wnt pathway.
Collapse
Affiliation(s)
- Yanli Shen
- Department of Pulmonary Medicine, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yan Yang
- Department of Pulmonary Medicine, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yan Zhao
- Department of Pulmonary Medicine, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Saiteer Nuerlan
- Department of Pulmonary Medicine, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yiyi Zhan
- Department of Pulmonary Medicine, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Chunling Liu
- Department of Pulmonary Medicine, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
46
|
Wang J, Zhu X, Jiang H, Ji M, Wu Y, Chen J. Cancer cell-derived exosome based dual-targeted drug delivery system for non-small cell lung cancer therapy. Colloids Surf B Biointerfaces 2024; 244:114141. [PMID: 39216444 DOI: 10.1016/j.colsurfb.2024.114141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024]
Abstract
Lung cancer is among most prevalent cancers in the world, in which non-small cell lung cancer (NSCLC) accounts for more than 85 % of all subtypes of lung cancers. NSCLC is often diagnosed at an advanced stage with a high mortality rate. Despite the demonstrated efficacy of chemotherapy in the treatment of NSCLC, the main drawback of current therapy is the lack of an effective drug-targeted delivery system, which may result in undesirable side effects during the clinical treatment. In this study, we construct a "dual-targeting" anti-cancer drug delivery platform by combining superparamagnetic iron oxide nanoparticles (SPIONs) with exosomes derived from NSCLC cells. We successfully promoted the targeted delivery of anti-drug doxorubicin (DOX) at the cellular levels by combining the homing targeted ability of exosomes with the magnetic targeted ability of SPIONs. Moreover, non-small cell lung cancer cell (NCI-h1299) tumor models were established. It was found that exosome-SPIONs (Exo-SPIONs) loaded with DOX exhibited optimal tumor tissue delivery and tumor suppression in the presence of an external magnetic field, and reduced the toxicity of the DOX to normal tissues. The constructed "dual-targeting" anti-cancer drug delivery platform holds promise for targeted chemotherapy for NSCLC.
Collapse
MESH Headings
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Exosomes/chemistry
- Exosomes/metabolism
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Doxorubicin/pharmacology
- Doxorubicin/chemistry
- Doxorubicin/administration & dosage
- Drug Delivery Systems
- Animals
- Cell Line, Tumor
- Mice
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/administration & dosage
- Cell Proliferation/drug effects
- Magnetic Iron Oxide Nanoparticles/chemistry
- Cell Survival/drug effects
- Mice, Nude
- Magnetite Nanoparticles/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Mice, Inbred BALB C
- Drug Screening Assays, Antitumor
Collapse
Affiliation(s)
- Jun Wang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinyi Zhu
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Huijun Jiang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Minghui Ji
- School of Nursing, Nanjing Medical University, Nanjing 211166, China
| | - Yuan Wu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China.
| | - Jin Chen
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
47
|
Li Z, Fan Y, Ma Y, Meng N, Li D, Wang D, Lian J, Hu C. Identification of Crucial Genes and Signaling Pathways in Alectinib-Resistant Lung Adenocarcinoma Using Bioinformatic Analysis. Mol Biotechnol 2024; 66:3655-3673. [PMID: 38142454 DOI: 10.1007/s12033-023-00973-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Alectinib, a second-generation anaplastic lymphoma kinase (ALK) inhibitor, has been shown to be effective for patients with ALK-positive non-small cell lung cancer (NSCLC). However, alectinib resistance is a serious problem worldwide. To the best of our knowledge, little information is available on its molecular mechanisms using the Gene Expression Omnibus (GEO) database. In this study, the differentially expressed genes (DEGs) were selected from the gene expression profile GSE73167 between parental and alectinib-resistant human lung adenocarcinoma (LUAD) cell samples. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) annotation enrichment analyses were conducted using Database for Annotation, Visualization and Integrated Discovery (DAVID). The construction of protein-protein interaction (PPI) network was performed to visualize DEGs. The hub genes were extracted based on the analysis of the PPI network using plug-in cytoHubba of Cytoscape software. The functional roles of the key genes were investigated using Gene Expression Profiling Interactive Analysis (GEPIA), University of Alabama at Birmingham Cancer (UALCAN), Gene Set Enrichment Analysis (GSEA), and Tumor Immune Estimation Resource (TIMER) analysis. The networks of kinase, miRNA, and transcription-factor targets of SFTPD were explored using LinkedOmics. The drug sensitivity analysis of SFTPD was analyzed using the RNAactDrug database. Results showed a total of 144 DEGs were identified. Five hub genes were extracted, including mucin 5B (MUC5B), surfactant protein D (SFTPD), deleted in malignant brain tumors 1 (DMBT1), surfactant protein A2 (SFTPA2), and trefoil factor 3 (TFF3). The survival analysis using GEPIA displayed that low expression of SFTPD had a significantly negative effect on the prognosis of patients with LUAD. GSEA revealed that low expression of SFTPD was positively correlated with the pathways associated with drug resistance, such as DNA replication, cell cycle, drug metabolism, and DNA damage repair, including mismatch repair (MMR), base excision repair (BER), homologous recombination (HR), and nucleotide excision repair (NER). The SFTPD expression was negatively correlated with the drug sensitivity of alectinib according to RNAactDrug database. The expression of SFTPD was further validated in parental H3122 cells and alectinib-resistant H3122 cells by quantitative reverse transcription PCR (RT-qPCR). In conclusion, our study found that the five hub genes, especially low expression of SFTPD, are closely related to alectinib resistance in patients with LUAD.
Collapse
Affiliation(s)
- Zhilong Li
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Yafeng Fan
- Respiratory Department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Yong Ma
- Thoracic Surgery Department II, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Nan Meng
- Department of Translational Medicine, ChosenMed Technology (Zhejiang) Co., Ltd, Beijing, 100176, China
| | - Dongbing Li
- Department of Translational Medicine, ChosenMed Technology (Zhejiang) Co., Ltd, Beijing, 100176, China
| | - Dongliang Wang
- Department of Translational Medicine, ChosenMed Technology (Zhejiang) Co., Ltd, Beijing, 100176, China
| | - Jianhong Lian
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China.
| | - Chengguang Hu
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China.
| |
Collapse
|
48
|
Wu T, Zeng L, Peng C, Zhao Z, Mu W, Wang S, Tan D. SNHG12 in cancer-associated fibroblast-derived extracellular vesicle induces macrophage-myofibroblast transition. Epigenomics 2024; 16:1415-1427. [PMID: 39568326 PMCID: PMC11622769 DOI: 10.1080/17501911.2024.2430166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024] Open
Abstract
AIMS To investigate mechanism of lncRNA SNHG12 induced macrophage-myofibroblast transition (MMT) in cancer-associated fibroblasts (CAFs)-derived extracellular vesicles (EVs) in non-small cell lung cancer (NSCLC). METHOD CAFs EVs were isolated from human NSCLC tissue and adjacent cancerous tissue (n = 3), and their morphology and particle size were evaluated. Macrophages and MMT cells with different phenotypes were detected, and the binding relationship of lncRNA SNHG12, miR-181a-5p, and Smad3 was verified. RESULT LncRNA SNHG12 derived from CAFs-EVs promoted the transformation of M2 macrophages into MMT. In addition, lncRNA-SNHG12 increased the expression of Smad3 which was significantly upregulated in MMT through sponge of miR-181a-5p. CONCLUSION LncRNA SNHG12 derived from CAFs-EV induced MMT in NSCLC.
Collapse
Affiliation(s)
- Tao Wu
- Cardiothoracic surgery department, Banan Hospital of Chongqing medical university, Chongqing, China
| | - Li Zeng
- Department of Pathology, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Chao Peng
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Zheng Zhao
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Weihao Mu
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Song Wang
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Deli Tan
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| |
Collapse
|
49
|
Xie Y, Chen H, Zhang X, Zhang J, Zhang K, Wang X, Min S, Wang X, Lian C. Integration of the bulk transcriptome and single-cell transcriptome reveals efferocytosis features in lung adenocarcinoma prognosis and immunotherapy by combining deep learning. Cancer Cell Int 2024; 24:388. [PMID: 39580462 PMCID: PMC11585238 DOI: 10.1186/s12935-024-03571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 11/10/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Efferocytosis (ER) refers to the process of phagocytic clearance of programmed dead cells, and studies have shown that it is closely related to tumor immune escape. METHODS This study was based on a comprehensive analysis of TCGA, GEO and CTRP databases. ER-related genes were collected from previous literature, univariate Cox regression was performed and consistent clustering was performed to categorize lung adenocarcinoma (LUAD) patients into two subgroups. Lasso regression and multivariate Cox regression analyses were used to construct ER-related prognostic features, and multiple immune infiltration algorithms were used to assess the correlation between the extracellular burial-related risk score (ERGRS) and tumor microenvironment (TME). And the key gene HAVCR1 was identified by deep learning, etc. Finally, pan-cancer analysis of the key genes was performed and in vitro experiments were conducted to verify the promotional effect of HAVCR1 on LUAD progression. RESULTS A total of 33 ER-related genes associated with the prognosis of LUAD were identified, and the prognostic signature of ERGRS was successfully constructed to predict the overall survival (OS) and treatment response of LUAD patients. The high-risk group was highly enriched in some oncogenic pathways, while the low-ERGRS group was highly enriched in some immune-related pathways. In addition, the high ERGRS group had higher TMB, TNB and TIDE scores and lower immune scores. The low-risk group had better immunotherapeutic response and less likelihood of immune escape. Drug sensitivity analysis revealed that BRD-K92856060, monensin and hexaminolevulinate may be potential therapeutic agents for the high-risk group. And ERGRS was validated in several cohorts. In addition, HAVCR1 is one of the key genes, and knockdown of HAVCR1 in vitro significantly reduced the proliferation, migration and invasion ability of lung adenocarcinoma cells. CONCLUSION Our study developed a novel prognostic signature of efferocytosis-related genes. This prognostic signature accurately predicted survival prognosis as well as treatment outcome in LUAD patients and explored the role of HAVCR1 in lung adenocarcinoma progression.
Collapse
Affiliation(s)
- Yiluo Xie
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| | - Xueying Zhang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Kai Zhang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Xinyu Wang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Shengping Min
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China.
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
50
|
Yeo H, Lee H, Park SM, Kang HN. Paeoniae radix overcomes resistance to EGFR-TKIs via aurora B pathway suppression in lung adenocarcinoma. Life Sci 2024; 357:123097. [PMID: 39362582 DOI: 10.1016/j.lfs.2024.123097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/04/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Targeted therapies using epidermal growth factor receptor (EGFR) inhibitors have markedly improved survival rates and quality of life for patients with EGFR-mutant lung adenocarcinoma (LUAD). Despite these advancements, resistance to EGFR inhibitors remains a significant challenge, limiting the overall effectiveness of the treatment. This study explored the synergistic effects of combining Paeoniae Radix (PR) with first-generation EGFR-tyrosine kinase inhibitors (TKIs), erlotinib and gefitinib, to overcome this resistance. Transcriptomic analysis of EGFR-mutant LUAD cell lines revealed that PR treatment could potentially reverse the gene signatures associated with resistance to EGFR-TKIs, primarily through the suppression of the Aurora B pathway. Experimental validation demonstrated that combining PR with erlotinib and gefitinib enhanced drug responsiveness by inhibiting Aurora kinase activity and inducing apoptosis in LUAD cells. Additionally, gene expression changes confirmed these combined effects, with the suppression of the Aurora B pathway and upregulation of the apoptotic pathway, which was accompanied by increased expression of multiple pro-apoptotic genes. Our findings contribute to the development of natural product-based therapeutic strategies to mitigate drug resistance in LUAD.
Collapse
Affiliation(s)
- Heerim Yeo
- College of Pharmacy, Chungnam National University, Republic of Korea
| | - Haeseung Lee
- College of Pharmacy, Busan National University, Republic of Korea
| | - Sang-Min Park
- College of Pharmacy, Chungnam National University, Republic of Korea.
| | - Han Na Kang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Republic of Korea.
| |
Collapse
|