1
|
Wang P, Zhang C, Wu P, Zhao Z, Sun N, Xue Q, Gao S, He J. Cell Death and Senescence-Based Molecular Classification and an Individualized Prediction Model for Lung Adenocarcinoma. MedComm (Beijing) 2025; 6:e70237. [PMID: 40443719 PMCID: PMC12122187 DOI: 10.1002/mco2.70237] [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: 01/12/2024] [Revised: 03/31/2025] [Accepted: 04/10/2025] [Indexed: 06/02/2025] Open
Abstract
The exploration of cell death and cellular senescence (CDS) in cancer has been an area of interest, yet a systematic evaluation of CDS features and their interactions in lung adenocarcinoma (LUAD) to understand tumor heterogeneity, tumor microenvironment (TME) characteristics, and patient clinical outcomes is previously uncharted. Our study characterized the activities and interconnections of 21 CDS features in 1788 LUAD cases across 15 cohorts, employing unsupervised clustering to categorize patients into three CDS subtypes with distinct TME profiles. The CDS index (CDSI), derived from principal component analysis, was developed to assess individual tumor CDS regulation patterns. Twelve CDSI core genes, enriched in proliferating T cells within the TME as per single-cell analysis, were identified and their functional roles and prognostic significance were validated. High CDSI correlated with improved overall survival in discovery cohort, four independent validation cohorts, and subgroup analysis. CDSI-low patients exhibited a favorable clinical response to immunotherapy and potential sensitivity to mitosis pathway drugs, while CDSI-high patients might benefit from drugs targeting ERK/MAPK and MDM2-p53 pathways. The clinical utility of CDSI was further validated using 9185 pan-cancer samples, demonstrating the broad relevance of our prediction model across various cancer types and its potential clinical implications for cancer management.
Collapse
Affiliation(s)
- Pan Wang
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chaoqi Zhang
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Peng Wu
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhihong Zhao
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nan Sun
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qi Xue
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shugeng Gao
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jie He
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Qu Y, Feng X, Chen H, Tan F, Shao A, Pang J, Xue Q, Zheng B, Zheng W, Ou Q, Gao S, Shao K. Multi-omics analyses reveal distinct molecular characteristics and transformation mechanisms of stage I-III micropapillary lung adenocarcinoma. J Pathol 2025; 266:204-216. [PMID: 40151900 DOI: 10.1002/path.6416] [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: 10/28/2024] [Revised: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 03/29/2025]
Abstract
The micropapillary (MIP) pattern is a high-grade histological subtype of lung adenocarcinoma (LUAD) with poor prognosis. In this study, surgically resected tumor samples from 101 patients with stage I-III MIP-LUAD (MIP ≥30%) were microdissected to separate MIP components from non-MIP components, all of which underwent RNA and DNA whole-exome sequencing (WES). The genomic and transcriptomic landscapes of MIP and non-MIP components within MIP-enriched tumor tissues demonstrated remarkable similarities, notably marked by high epidermal growth factor receptor (EGFR) alteration frequencies. However, when compared to MIP-naïve LUAD tissues, MIP components showed higher chromosomal instability and revealed 18 enriched alterations, encompassing EGFR mutations, EGFR amplifications, and CDKN2A/CDKN2B deletions, which all linked to upregulation of cell proliferation pathways and downregulation of immune pathways. Shared mutations were observed in 97.8% (91/93) of patients with paired DNA WES data for MIP and non-MIP components within the same tissues, suggesting a common origin. The recurrence-free survival analysis identified MACF1, PCLO, ADGRV1, and Fanconi Anemia pathway mutations as negative indicators. In all, we conducted an in-depth analysis of the molecular characteristics and transformation mechanisms of MIP-LUAD, employing microdissection techniques to investigate the genomic and transcriptomic levels within a substantial cohort, providing insights for precision medicine of this aggressive cancer subtype. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yang Qu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Hanlin Chen
- Nanjing Geneseeq Technology Inc, Nanjing, PR China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Anqi Shao
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Jiaohui Pang
- Nanjing Geneseeq Technology Inc, Nanjing, PR China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Wei Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Qiuxiang Ou
- Nanjing Geneseeq Technology Inc, Nanjing, PR China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Kang Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| |
Collapse
|
3
|
Rueda-Zarazua B, Gutiérrez H, García-Ortiz H, Orozco L, Ramírez-Martínez G, Jiménez-Alvarez L, Bolaños-Morales FV, Zuñiga J, Ávila-Moreno F, Melendez-Zajgla J. A Pilot Study: Contrasting Genomic Profiles of Lung Adenocarcinoma Between Patients of European and Latin American Ancestry. Int J Mol Sci 2025; 26:4865. [PMID: 40430005 PMCID: PMC12111962 DOI: 10.3390/ijms26104865] [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: 03/19/2025] [Revised: 05/06/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Lung cancer remains as the leading cause of cancer mortality worldwide. However, while current evidence suggests the existence of genomic differences between populations, indicating different risk factors associated with population-level genetic backgrounds, most studies have concentrated on populations of European ancestry, and more research is needed on non-European populations. We analyzed whole-exome sequencing data from 25 Mexican lung adenocarcinoma patients and compared them with a TCGA-PanCancer cohort enriched with patients of European ancestry as reference. Clinically relevant germline variants in cancer susceptibility genes are more frequent in our cohort (32% vs. 6.4%) than in the reference. Several mutational signatures (SBS32, SBS85, SBS12, SBS19) occurred at significantly higher frequencies in the Mexican cohort compared to the reference (p < 0.0001). Interestingly, the smoking-associated signature SBS4, present in 67.6% of smokers in the reference cohort, was absent in smoking Mexican patients (p < 0.01656). Somatic variant frequencies in SLC36A4 (20%; p < 0.00002), AP1S1 (8%; p < 0.00002), and TP53 (16%; p = 0.00005) showed significant differences from the European reference cohort. We demonstrate that all these observed biases were independent of the sample size. This study uncovers distinct genomic biases in lung cancer carcinogenesis in this population, compared to a European ancestry reference population, suggesting implications for precision medicine strategies in Latin American populations.
Collapse
Affiliation(s)
- Bertha Rueda-Zarazua
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | | | - Humberto García-Ortiz
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico; (H.G.-O.); (L.O.)
| | - Lorena Orozco
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico; (H.G.-O.); (L.O.)
| | - Gustavo Ramírez-Martínez
- Laboratorio de Inmunología y Genética, Instituto Nacional de Enfermedades Respiratorias, Mexico City 14080, Mexico; (G.R.-M.); (L.J.-A.); (J.Z.)
| | - Luis Jiménez-Alvarez
- Laboratorio de Inmunología y Genética, Instituto Nacional de Enfermedades Respiratorias, Mexico City 14080, Mexico; (G.R.-M.); (L.J.-A.); (J.Z.)
| | | | - Joaquín Zuñiga
- Laboratorio de Inmunología y Genética, Instituto Nacional de Enfermedades Respiratorias, Mexico City 14080, Mexico; (G.R.-M.); (L.J.-A.); (J.Z.)
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City 14380, Mexico
| | - Federico Ávila-Moreno
- Lung Diseases and Functional Epigenomics Laboratory (LUDIFE), Biomedicine Research Unit (UBIMED), Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico;
- Research Tower, Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Jorge Melendez-Zajgla
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| |
Collapse
|
4
|
Wang Y, Liu N, Xu C, Wang J, Dong L, Yang S, Jiang J. Nebulized inhalation of extracellular vesicles containing SPOCK2 suppresses lung adenocarcinoma progression via MAPK inhibition. Discov Oncol 2025; 16:797. [PMID: 40382517 PMCID: PMC12085455 DOI: 10.1007/s12672-025-02626-9] [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: 11/15/2024] [Accepted: 05/08/2025] [Indexed: 05/20/2025] Open
Abstract
Aberrant expression of SPARC/osteonectin, cwcv and kazal-like domains proteoglycan 2 (SPOCK2) plays a role in the development and progression of several human cancers. However, the importance of its expression and function in lung adenocarcinoma (LUAD) remains unclear. The present study aimed to elucidate the role of SPOCK2 in the growth of LUAD and propose a novel therapeutic insight for LUAD through SPOCK2. SPOCK2 protein expression was significantly reduced in LUAD tissues and cells by Immunohistochemical assay and Western blot. CCK-8, colony formation, and Transwell assays were used to demonstrate that SPOCK2 overexpression inhibited both proliferation and migration of LUAD cells in vitro. This inhibition of tumor growth was further confirmed by a LUAD xenograft mouse model in vivo. To explore downstream target signal of SPOCK2 in LUAD, RNA transcriptome sequencing was performed and enrichment analysis showed an association between SPOCK2 expression and the MAPK pathway. Furthermore, HEK293T cells were modified with SPOCK2, and extracellular vesicles (EVs) containing SPOCK2 (SPOCK2-EVs) were collected through ultra-high-speed centrifugation. Interestingly, co-culture with SPOCK2-EVs significantly increased SPOCK2 levels within LUAD cells. Furthermore, SPOCK2-EVs effectively inhibited LUAD growth in vitro and in vivo studies. Because directly injecting SPOCK2-EVs into tumors presents challenges for internal organs, we investigated the efficacy of nebulized SPOCK2-EVs for LUAD treatment. Consistent with our findings from intratumoral injection, nebulized inhalation of SPOCK2-EVs resulted in significant inhibition of LUAD growth. These results strongly suggest that SPOCK2 released by HEK293T-EVs can effectively inhibit LUAD tumor growth and hold promise for future clinical translation in cancer therapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, No. 9 Chongwen Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Ningning Liu
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Chuanqin Xu
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Jing Wang
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Liyang Dong
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu, People's Republic of China.
| | - Shuang Yang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, No. 9 Chongwen Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China.
- Center for Clinical Mass Spectrometry, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
| | - Junhong Jiang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, No. 9 Chongwen Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
5
|
Mao S, Li Q, Yang Y, Liu Z, Zhang L. Potential Crosstalk Between ANXA1+ Epithelial Cells and FABP4+ TAM Cells of Ferroptosis-Related Molecular Clusters Promotes an Immunosuppressive Microenvironment in Non-Small Cell Lung Cancer. Mol Carcinog 2025; 64:936-950. [PMID: 40040274 DOI: 10.1002/mc.23899] [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: 09/25/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 03/06/2025]
Abstract
The tumor microenvironment (TME) affects tumor initiation, invasion, metastasis, and therapies. Recently, increasing evidence has demonstrated that ferroptosis plays important regulatory roles in tumourigenesis and progression. It is unclear how ferroptosis affects non-small cell lung cancer (NSCLC) progression by remodeling the TME. In this study, the single-cell RNA sequencing (scRNA-seq) data (85,562 cells, n = 18) were employed to reveal the heterogeneity of ferroptosis activation in NSCLC, and identified six ferroptosis-related molecular clusters. We found that ANXA1+ epithelial and FABP4 + TAM subpopulations were key factors in lung cancer progression and TME remodeling. In addition, the cell-cell communication analysis showed that ANXA1-FPR2/FPR1 receptor-ligand pair contributed to the formation of an immunosuppressive TME. Furthermore, we established a novel signature based on ferroptosis-related molecular clusters, and the risk score model may predict survival and response to immunotherapy. We also found that compared with responder, the expression of ANXA1 and FABP4 is higher in progressor, which indicating a higher expression of ANXA1 and FABP4 was associated with a worse response to immunotherapy. Therefore, we concluded that the molecular clusters associated with ferroptosis served as potential prognostic markers and therapeutic targets for NSCLC patients.
Collapse
Affiliation(s)
- Shengqiang Mao
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingyan Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Yang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhiqiang Liu
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
6
|
Mao S, Chen L, Li Q, Zhang L, Zhao H, Lin Y. Unveiling hypoxia-related prognostic and immunotherapeutic biomarkers in lung adenocarcinoma through single-cell and bulk RNA sequencing: Including insights into PGF. Int J Biol Macromol 2025; 309:143056. [PMID: 40228772 DOI: 10.1016/j.ijbiomac.2025.143056] [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: 10/21/2024] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Hypoxia plays a crucial role in lung adenocarcinoma (LUAD) proliferation and metastasis. However, the mechanisms underlying the interaction between the hypoxic microenvironment and immune resistance remain unclear. In this study, single-cell RNA sequencing (scRNA-seq) data from 15 LUAD patients were used to evaluate the complexity and heterogeneity of tumor microenvironment (TME). We identified new subtypes associated with advanced LUAD, including epithelial cells, fibroblasts, and myeloid cells. Furthermore, we found that the cell subtype module 3 (AGER, TIMP3) of epithelial cells exhibited higher hypoxia scores in advanced LUAD. Meanwhile, we also observed that RSG5 + fibroblast, AOPE+macrophage, S100B + macrophage, CCL17 + macrophage, and HLA-DRB5 + macrophage cells exhibited higher hypoxia scores in advanced LUAD patients. Moreover, spatial transcriptomic analysis revealed that with the gradual decrease of hypoxia score, the cell type score also gradually decreased. Cell communication analysis identified critical receptor-ligand pairs, which were associated with the activation of the PD-1/PD-L1 pathway. Finally, we developed a novel prognostic signature based on hypoxia-related molecular clusters, which possessed predictive power for both prognosis and immunotherapy response. The experimental results confirmed that hypoxia-related genes play a significant role in driving LUAD progression. In conclusion, our study provides valuable insights into the hypoxic and immunosuppressive tumor microenvironment, which serve as a potential prognostic marker and therapeutic target for LUAD.
Collapse
Affiliation(s)
- Shengqiang Mao
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lu Chen
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingyan Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huachang Zhao
- Department of Respiratory and Critical Care Medicine, The Fourth People's Hospital of Chengdu, No.8 Huli-West 1st-Alley, Jin-Niu District, Chengdu, China; School of Life Science and Technology, University of Electronic Science and Technology of China, Qingshuihe Campus: No.2006, Xiyuan Ave, West Hi-Tech Zone, Chengdu, China.
| | - Yidan Lin
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
7
|
Lin YD, Li HJ, Hong HZ, Qi YF, Li YY, Yang XN, Wu YL, Zhong WZ. Genomic profiling of aggressive pathologic features in lung adenocarcinoma. Lung Cancer 2025; 203:108460. [PMID: 40179539 DOI: 10.1016/j.lungcan.2025.108460] [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: 05/30/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION Pathologic features involving LVI (lympho-vascular invasion), PNI (perineural invasion), STAS (spread through air spaces), and Grade 3 pattern (from the International Association for the Study of Lung Cancer grading system) are related to having an aggressive phenotype and linked to poor prognosis. However, few studies have conducted in-depth analyses of these features simultaneously with genomic profiling. METHODS A total of 1559 sequencing of adenocarcinoma samples were included in the common driver mutations analysis, 1306 samples were brought into genomic mapping analysis. OncoSG's East Asian ancestry dataset was implemented for Tumor-Node-Metastasis-Biomarker (TNMB) classification and prognostic assessment. RESULTS EGFR was more significantly prevalent in LVI negativity (P = 0.021), STAS negativity (P = 0.002), and moderate grade (P < 0.001). ALK was significantly interrelated with LVI (P = 0.028), STAS (P < 0.001), and poor grade (P < 0.001); ROS1 and STAS positivity (P = 0.031), poor grade (P = 0.016) were significantly related. KRAS (P = 0.003) and BRAF-V600E (P = 0.002) were only significantly intertwined with poor grade. Apart from common driver mutations, TP53, CHEK2, KEAP1, PTEN, RB1, NF1 were significantly enriched in LVI samples (P < 0.05). TP53, PTEN, CTNNB1, HGF, NF1 were more prominent in STAS (P < 0.01). TP53, LRP1B, NF1 were significantly more prevalent in Grade 3 pattern (P < 0.001). The mixture of STK11, PTEN, and TOP2A generated by exclusive mutations may be a potential predictor of TNMB categorization towards survival. The HR of stage II compared I of TNMB was 2.28 (95 % CI 1.36-3.86, P < 0.001), while stage III compared II was 1.95 (95 % CI 1.04-3.21, P = 0.031). CONCLUSIONS This analysis demonstrated the correlation of pathologic features with common driver mutations, key mutations and canonical oncogenic signaling pathways. The data highlighted the similarities and differences among these features horizontally, and provide new insights in TNMB classification and prognostic assessment.
Collapse
Affiliation(s)
- Yi-Duo Lin
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hong-Ji Li
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hui-Zhao Hong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Fan Qi
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yun-Yi Li
- School of Software Engineering, South China University of Technology, Guangzhou, China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Liu B, Tao W, Zhou X, Xu LD, Luo Y, Yang X, Min Q, Huang M, Zhu Y, Cui X, Wang Y, Gong T, Zhang E, Huang YS, Chen W, Yan S, Wu N. Multi‑omics analysis identifies different molecular subtypes with unique outcomes in early-stage poorly differentiated lung adenocarcinoma. Mol Cancer 2025; 24:129. [PMID: 40312720 PMCID: PMC12044723 DOI: 10.1186/s12943-025-02333-7] [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: 02/22/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025] Open
Abstract
INTRODUCTION Early-stage poorly differentiated lung adenocarcinoma (LUAD) is plagued by a high risk of postoperative recurrence, and its prognostic heterogeneity complicates treatment and surveillance planning. We conducted this integrative multi-omics study to identify those patients with a truly high risk of adverse outcomes. METHODS Whole-exome, RNA and whole methylome sequencing were carried out on 101 treatment-naïve early-stage poorly differentiated LUADs. Integrated analyses were conducted to disclose molecular characteristics and explore molecular subtyping. Functional validation of key molecules was carried out through in vitro and in vivo experiments. RESULTS Recurrent tumors exhibited significantly higher ploidy (p = 0.024), the fraction of the genome altered (FGA, p = 0.042), and aneuploidy (p < 0.05) compared to non-recurrent tumors, as well as a higher frequency of CNVs. Additionally, recurrent tumors showed hypomethylation at both the global level and in CpG island regions. Integrative transcriptomic and methylation analyses identified three molecular subtypes (C1, C2, and C3), with the C1 subtype presenting the worst prognosis (p = 0.024). Although frequently mutated genes showed similar mutation frequencies across the three subtypes, the C1 subtype exhibited the highest tumor mutation burden (TMB), mutant-allele tumor heterogeneity (MATH), aneuploidy, and HLA loss of heterozygosity (HLA-LOH), along with relatively lower immune cell infiltration. Furthermore, GINS1 and CPT1C were found to promote LUAD progression, and their high expression correlated with a poor prognosis. CONCLUSIONS This multi-omics study identified three integrative subtypes with distinct prognostic implications, paving the way for more precise management and postoperative monitoring of early-stage poorly differentiated LUAD.
Collapse
Affiliation(s)
- Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wei Tao
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xuantong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Li-Di Xu
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yanrui Luo
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Miao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xinrun Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yaqi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tongyang Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Enli Zhang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yu S Huang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Weizhi Chen
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Nan Wu
- State Key Laboratory of Molecular Oncology, Frontiers Science Center for Cancer Integrative Omics, Department of Thoracic Surgery II, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan, China.
| |
Collapse
|
9
|
Turongkaravee S, Nathisuwan S, Baisamut T, Meanwatthana J. Cost-Utility Analysis of Genomic Profiling in Directing Targeted Therapy in Advanced NSCLC in Thailand. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2025; 23:479-491. [PMID: 39918787 PMCID: PMC12052874 DOI: 10.1007/s40258-025-00950-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/19/2025] [Indexed: 05/06/2025]
Abstract
BACKGROUND Sequential next-generation sequencing (NGS) testing has demonstrated cost-effectiveness in guiding targeted therapy with tyrosine kinase inhibitors (TKIs) for advanced non-small cell lung cancer (aNSCLC) in developed countries. However, its cost-effectiveness in developing countries remains uncertain. OBJECTIVE The aim was to conduct a cost-utility analysis comparing sequential NGS testing with the current approach of single epidermal growth factor receptor (EGFR) testing combined with first-line targeted therapy, as implemented under Thailand's Universal Health Coverage scheme for aNSCLC. METHOD Hybrid decision tree and Markov models were developed to estimate the lifetime costs and quality-adjusted life years (QALYs) associated with each strategy. The models simulate cohorts of aNSCLC patients who receive platinum-based chemotherapy or TKIs based on identified gene alterations. Patients enter the model at 60 years of age. The incremental cost-effectiveness ratio (ICER) was computed from a societal perspective. The analysis employed a lifetime horizon and discounted costs and outcomes at a rate of 3%. Furthermore, uncertainty and scenario analyses were conducted. FINDINGS A sequential NGS testing strategy could identify an additional 19% of patients with biomarker-positive findings who subsequently received biomarker-driven targeted therapy compared to a single EGFR testing strategy. The number needed to screen to identify a single gene mutation and administer first-line TKI was six for the sequential NGS testing strategy. Compared to the single EGFR testing, the ICER of the sequential NGS testing strategy was 1,851,150 THB/QALY (US$51,335). At a willingness-to-pay threshold of 160,000 THB/QALY (US$4437), the single EGFR testing strategy demonstrated 100% cost-effectiveness. In contrast, the sequential NGS testing was not deemed cost-effective. The sensitivity of the ICER was influenced by the overall survival rates associated with anaplastic lymphoma kinase (ALK) inhibitors and platinum-based chemotherapy. INTERPRETATION Sequential NGS testing identified a greater number of patients with aNSCLC eligible for targeted therapies, resulting in improved survival rates and enhanced QALYs compared to single EGFR testing. However, in the context of Thailand, sequential NGS testing was not cost-effective. The single EGFR testing strategy emerged as the most cost-effective option for guiding first-line targeted therapy.
Collapse
Affiliation(s)
- Saowalak Turongkaravee
- Social and Administrative Pharmacy Division, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Surakit Nathisuwan
- Division of Clinical Pharmacy, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Rd, Phayathai, Ratchathewi, Bangkok, 10400, Thailand
| | - Thanyanan Baisamut
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jennis Meanwatthana
- Division of Clinical Pharmacy, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Rd, Phayathai, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
10
|
Yu J, Kong X, Feng Y. Tumor microenvironment-driven resistance to immunotherapy in non-small cell lung cancer: strategies for Cold-to-Hot tumor transformation. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:21. [PMID: 40342732 PMCID: PMC12059482 DOI: 10.20517/cdr.2025.14] [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: 01/14/2025] [Revised: 03/19/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Non-small cell lung cancer (NSCLC) represents a formidable challenge in oncology due to its molecular heterogeneity and the dynamic suppressive nature of its tumor microenvironment (TME). Despite the transformative impact of immune checkpoint inhibitors (ICIs) on cancer therapy, the majority of NSCLC patients experience resistance, necessitating novel approaches to overcome immune evasion. This review highlights shared and subtype-specific mechanisms of immune resistance within the TME, including metabolic reprogramming, immune cell dysfunction, and physical barriers. Beyond well-characterized components such as regulatory T cells, tumor-associated macrophages, and myeloid-derived suppressor cells, emerging players - neutrophil extracellular traps, tertiary lymphoid structures, and exosomal signaling networks - underscore the TME's complexity and adaptability. A multi-dimensional framework is proposed to transform cold, immune-excluded tumors into hot, immune-reactive ones. Key strategies include enhancing immune infiltration, modulating immunosuppressive networks, and activating dormant immune pathways. Cutting-edge technologies, such as single-cell sequencing, spatial transcriptomics, and nanomedicine, are identified as pivotal tools for decoding TME heterogeneity and personalizing therapeutic interventions. By bridging mechanistic insights with translational innovations, this review advocates for integrative approaches that combine ICIs with metabolic modulators, vascular normalizers, and emerging therapies such as STING agonists and tumor vaccines. The synergistic potential of these strategies is poised to overcome resistance and achieve durable antitumor immunity. Ultimately, this vision underscores the importance of interdisciplinary collaboration and real-time TME profiling in refining precision oncology for NSCLC, offering a blueprint for extending these advances to other malignancies.
Collapse
Affiliation(s)
- Jinglu Yu
- Institute of Integrated Chinese and Western Medicine, PuDong Traditional Chinese Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201200, China
- Institute of Respiratory Medicine, PuDong Traditional Chinese Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201200, China
| | - Xiaoni Kong
- Institute of Integrated Chinese and Western Medicine, PuDong Traditional Chinese Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201200, China
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu Feng
- Institute of Integrated Chinese and Western Medicine, PuDong Traditional Chinese Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201200, China
| |
Collapse
|
11
|
Wang L, Lin F, Yuan J, Wu X, Zhong Y, Li S, Lv Y. FAM207A acts as a novel and potential biomarker in lung adenocarcinoma and shapes the immunesuppressive tumor microenvironment. Clin Exp Med 2025; 25:125. [PMID: 40259152 PMCID: PMC12011971 DOI: 10.1007/s10238-025-01657-4] [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: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/23/2025]
Abstract
The expression of Family with sequence similarity 207 member A( FAM207A) is closely related to the development, growth, and progression of various cancers. However, extensive research into its biological functions remains unexplored. In this study, we conducted a comprehensive biological information analysis of the Lung adenocarcinoma (LUAD) dataset to elucidate the foundational mechanisms underlying FAM207A's role in tumor development. The expression and clinical information of LUAD patients for FAM207A were extracted from the Cancer Genome Atlas (TCGA). Using Western blot, we assessed the expression levels of relevant proteins in LUAD cells and human lung epithelial cells. Subsequently, we employed Cox regression analysis to evaluate the prognostic significance of FAM207A in LUAD, along with gene set enrichment analysis (GSEA) to explore its potential biological functions and interactions with FAM207A's immune microenvironment. Finally, in vitro experiments confirmed that FAM207A significantly influences the proliferation and migration of LUAD cells. The results indicate that FAM207A mRNA and protein expression levels in LUAD tissues and cell are significantly elevated. Additionally, FAM207A high expression is significantly associated with a shorter overall survival (OS) and more advanced pathological stages. Furthermore, FAM207A expression is significantly linked to the expression of immunogenic markers in the LUAD tumor microenvironment. Gene set and KEGG enrichment analyses revealed that FAM207A is primarily associated with genes involved in adhesion and immune signaling pathways. Additionally, in vitro experiments demonstrated that FAM207A can effectively promote the proliferation and migration of LUAD cells. Our findings revealed that FAM207A is overexpressed in LUAD and is linked to a poor prognosis. Our study demonstrates the potential of FAM207A as an immunotherapeutic and predictive biomarker in LUAD.
Collapse
Affiliation(s)
- Lu Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feihong Lin
- Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jixiang Yuan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yushan Zhong
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Li
- Shandong Provincial Hospital, Jinan, Shandong, China.
| | - Ya Lv
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
12
|
Tajè R, Gallina FT, Caterino M, Forcella D, Patirelis A, Alessandrini G, Buglioni S, Cecere FL, Fusco F, Cappelli F, Melis E, Visca P, Cappuzzo F, Ambrogi V, Vidiri A. Molecular characterization of early-stage lung adenocarcinoma presenting as subsolid nodules in a real-life European cohort. BMC Cancer 2025; 25:647. [PMID: 40205411 PMCID: PMC11983824 DOI: 10.1186/s12885-025-13998-0] [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: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVES Subsolid nodules emerged as frequent radiological variants of lung adenocarcinoma. Radiological features including solid-component prevalence and larger tumour dimensions prompt tumoral invasiveness guiding prognosis and management. Thus, we aimed to clarify the molecular grounds that dictate these radiological appearances and clinical behaviour in a real-life European-cohort. Additionally, following the growing interest toward targeted-therapies in early-stage diseases, we aimed to present real-life epidemiological data of actionable mutations in these patients. METHODS In this retrospective single-centre study, targeted next-generation sequencing was performed continuatively in all the resected subsolid lung adenocarcinomas in the period between May 2016 and December 2023. Clinico-radiological data were collected. The genetic landscape of our real-life European subsolid adenocarcinoma population is defined. Common and actionable mutations (frequency > 5%) relation to key clinico-radiological features are evaluated. RESULTS Overall, 156 subsolid adenocarcinomas were analysed. KRAS-mutations, mostly KRAS p.G12C, were the most prevalent followed by EGFR, including 25% uncommon EGFR-mutations, TP53 and MET mutations. Amongst the clinico-radiological variables, KRAS-mutations and KRAS p.G12C-mutation were associated to smoking history (≥ 20 pack/years), aggressive histologic subtype and higher consolidation-to-tumor ratio (CTR). Moreover, KRAS-mutated nodules had faster tumour-doubling-time. Conversely, EGFR-mutations were associated to female sex and lower CTR. The latter not being confirmed in common EGFR-mutations. Additionally, in common EGFR-mutated nodules, aggressive histological components were rarer. CONCLUSION Our study presents the molecular profile of subsolid lung adenocarcinoma in a real-life European-cohort. KRAS-mutations were the most prevalent, and were related to smoking history, higher CTR and faster growth. Conversely, common EGFR-mutations were rarer than expected and unrelated to smoking history and radiological features.
Collapse
Affiliation(s)
- Riccardo Tajè
- Doctoral School of Microbiology, Immunology, Infectious Diseases and Transplants, MIMIT, University of Rome "Tor Vergata", Rome, Italy
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Filippo Tommaso Gallina
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy.
- Tumor Immunology and Immunotherapy Unit, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy.
| | - Mauro Caterino
- Department of Radiology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Daniele Forcella
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Alexandro Patirelis
- Doctoral School of Microbiology, Immunology, Infectious Diseases and Transplants, MIMIT, University of Rome "Tor Vergata", Rome, Italy
- Department of Thoracic Surgery, Tor Vergata University, Rome, Italy
| | - Gabriele Alessandrini
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | | | - Francesca Fusco
- Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Federico Cappelli
- Department of Radiology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Enrico Melis
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Paolo Visca
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Federico Cappuzzo
- Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Vincenzo Ambrogi
- Department of Thoracic Surgery, Tor Vergata University, Rome, Italy
| | - Antonello Vidiri
- Department of Radiology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| |
Collapse
|
13
|
Goto Y, Watanabe S, Yanagimura N, Arita M, Sato M, Nozaki K, Tanaka T, Saida Y, Maemondo M, Kobayashi K, Hagiwara K, Kikuchi T. The diagnostic value of comprehensive next-generation sequencing for genetic mutations in suspected lung cancer cases with negative pathological cytology. Lung Cancer 2025; 202:108505. [PMID: 40117845 DOI: 10.1016/j.lungcan.2025.108505] [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: 01/26/2025] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION In recent clinical practice, driver gene mutations have been tested using multiplex PCR or next-generation sequencing (NGS), which help determine treatment strategies for non-small cell lung cancer (NSCLC). We developed a new analysis system, the Mutation Investigator using Next-era Sequencer (MINtS), using NGS, which allows for the detection of gene mutations even in cytology specimens with low tumor cell content. Due to its high sensitivity, MINtS has the potential to detect gene mutations even in specimens that are pathologically negative for cancer. In the present study, we examined the utility of MINtS-based mutation detection in cytology-negative specimens. METHODS We retrospectively analyzed the data of patients who were enrolled in the NEJ021A study, which was a prospective observational study investigating the performance of MINtS. Although NEJ021A was a multicenter study, we included only patients enrolled at Niigata University Medical and Dental Hospital. RESULTS Cytology specimens from 486 patients with suspected lung cancer were analyzed using MINtS. Among the cytology-positive cases, driver gene mutations were detected in 37.3 % (93/249) of patients, whereas in cytology-negative cases, driver gene mutations were detected in 20.2 % (47/233) of patients using MINtS. Of the 47 patients whose specimens were cytology-negative and MINtS-positive, 42 were histologically or clinically diagnosed with NSCLC and received treatment. CONCLUSIONS Even in patients without a pathological diagnosis of lung cancer, MINtS can identify driver gene mutations, which can be useful for guiding subsequent treatment decisions.
Collapse
Affiliation(s)
- Yuka Goto
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Naohiro Yanagimura
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masashi Arita
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Miyuki Sato
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Koichiro Nozaki
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomohiro Tanaka
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yu Saida
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Makoto Maemondo
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kunihiko Kobayashi
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Saitama, Japan
| | - Koichi Hagiwara
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
14
|
Mével-Aliset M, Radu AG, Allard J, Blanchet S, Montellier E, Hainaut P, Rossignol R, Torch S, Orsi GA, Thibert C. Transcriptional regulation by LKB1 in lung adenocarcinomas: Exploring oxidative stress, neuroglial and amino acid signatures. Biochem Biophys Res Commun 2025; 755:151571. [PMID: 40043609 DOI: 10.1016/j.bbrc.2025.151571] [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: 10/19/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Lung adenocarcinoma (LUAD) is one of the most prevalent cancer types worldwide and has one of the poorest survival rates. Understanding its developpment is crucial for improving diagnosis, prognosis, and treatment. A key factor in LUAD is the frequent loss-of-function mutations in LKB1/STK11, a kinase that regulates metabolism. These mutations are linked to increased metastasis and worse clinical outcomes. In this study, we analyzed gene expression data from LUAD patients to explore how LKB1 mutations affect cancer behavior. We found that LKB1 mutations in KRAS-driven LUAD lead to widespread gene downregulation. By integrating avalaible protein interaction data, mass spectrometry analysis of LKB1 nuclear partners, and co-immunoprecipitations experiments, we identified BRG1, a chromatin activator and subunit of the BAF complex, as a nuclear partner of LKB1. Further analysis suggested that LKB1 mutations may impair BRG1 activity, disrupting chromatin regulation and gene expression. Notably, LUAD patients with mutated LKB1 showed gene expression patterns indicative of oxidative stress, defective neuronal-glial and neuroinflammation programs, and altered amino acid homeostasis. These changes resemble the roles LKB1 plays in neural crest stem cells, suggesting that LKB1 may reduce tumor aggressiveness in LUAD by maintaining a developmental gene expression program.
Collapse
Affiliation(s)
- Marie Mével-Aliset
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Anca G Radu
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Jordan Allard
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Sandrine Blanchet
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Emilie Montellier
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Pierre Hainaut
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Rodrigue Rossignol
- INSERM U1211, Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; CELLOMET, Functional Genomics Center (CGFB), 146 rue Léo Saignat, 33076, Bordeaux, France
| | - Sakina Torch
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Guillermo A Orsi
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics of Regeneration and Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Chantal Thibert
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team "Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer", Institute for Advanced Biosciences, 38000, Grenoble, France.
| |
Collapse
|
15
|
Tavernari D, Borgeaud M, Liu X, Parikh K, Le X, Ciriello G, Addeo A. Decoding the Clinical and Molecular Signatures of EGFR Common, Compound, and Uncommon Mutations in NSCLC: A Brief Report. J Thorac Oncol 2025; 20:500-506. [PMID: 39694414 DOI: 10.1016/j.jtho.2024.12.012] [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: 10/29/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION EGFR mutations are key oncogenic drivers in lung adenocarcinoma (LUAD), predominantly affecting Asian, nonsmoking, and female populations. Although common mutations, such as exon 19 deletions and L858R, respond well to tyrosine kinase inhibitors (TKIs), uncommon EGFR mutations and compound variants exhibit variable treatment responses. This study aims to compare clinical characteristics and molecular profiles of patients with common, uncommon, and compound EGFR mutations, assessing their implications for therapy outcomes. METHODS We analyzed a multi-cohort genomic dataset of 19,163 patients with LUAD (5,212 with EGFR mutations), categorizing mutations into common, uncommon, and compound classes. Patient demographics, mutational signatures, and tumor microenvironment factors were assessed, with particular attention to smoking status and concomitant alterations in KRAS and TP53. Treatment outcomes were analyzed by time under treatment as a surrogate measure of TKI efficacy. RESULTS Uncommon EGFR mutations, comprising 8.9% of EGFR-altered cases, were significantly more frequent among smokers and associated with tobacco-related mutational signatures. Compared with common EGFR-mutant cases, tumors harboring uncommon EGFR mutations reported higher rates of EGFR amplifications, KRAS, and TP53 mutations. Uncommon mutations also exhibited higher tumor mutational burden and distinct transcriptional profiles linked to cell cycle activity. Median time on treatment with TKIs was notably shorter in patients with uncommon mutations (4.1 mo) than those with common and compound mutations (10.9 mo and 12.4 mo, respectively). CONCLUSIONS This study underscores the clinical and molecular heterogeneity of EGFR mutation classes in LUAD, highlighting the unique profile of uncommon mutations, particularly their association with smoking and co-mutations in KRAS and TP53. Comprehensive molecular testing, including next-generation sequencing, is crucial to identify these uncommon mutations and inform therapeutic decisions. Further investigation into the role of immunotherapy in patients with uncommon EGFR mutations is warranted given the tobacco-related molecular signatures and high tumor mutational burden associated with this subgroup.
Collapse
Affiliation(s)
- Daniele Tavernari
- Department of Computational Biology, University of Lausanne (UNIL), 1011 Lausanne, Vaud, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maxime Borgeaud
- Oncology Service, University Hospital Geneva, Geneva, Switzerland
| | - Ximeng Liu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Giovanni Ciriello
- Department of Computational Biology, University of Lausanne (UNIL), 1011 Lausanne, Vaud, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Alfredo Addeo
- Oncology Service, University Hospital Geneva, Geneva, Switzerland.
| |
Collapse
|
16
|
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
|
17
|
Li J, Li X, Guo H, Zhou S, Ding H, Li B, Li W, Zhang B, Zou Y. Multi-gene panel sequencing reveals the relationship between driver gene mutation and clinical characteristics in lung adenocarcinoma. Discov Oncol 2025; 16:274. [PMID: 40053265 DOI: 10.1007/s12672-025-02008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Testing of multiple cancer related genes using next-generation sequencing (NGS) has been widely used for personalized precision medicine of cancer. Integrated analysis of those NGS data and clinical data has offered new opportunities for investigating the relationship between driver genes' mutations and clinical characteristics in large cohorts. This study aims to explore the mutational landscape and its association with clinical features in a lung adenocarcinoma (LUAD) cohort. METHODS Tumor tissues from 132 LUAD patients were subjected to customized 30 genes targeted next-generation sequencing. Somatic mutations of the 30 genes were identified and annotated. Statistical analysis was performed to determine the cooccurrence of mutations of different driver genes and the association relationships between gene mutation and clinical features including gender and age. RESULTS A total of 96.97% (128/132) of LUAD patients experienced genetic mutations. EGFR had the highest mutation rate (81, 61.36%) among the 30 genes, followed by TP53 (80, 60.61%), BRAF (30, 22.73%), KRAS (21, 15.91%) and ROS1 (21, 15.91%). The L858R substitution and exon19 deletion were the predominant mutations of EGFR, accounting for 82.71% of EGFR-mutated patients. The 27 mutation sites of EGFR were mainly located in the tyrosine kinase catalytic domain (22/27, 81.48%). Mutations of SDHA (p < 0.01), ERBB2 (p < 0.01), and ESR1 (p < 0.05) were negatively correlated with age, and mutations of NF1 (p < 0.01), KRAS (p < 0.01), and TP53 (p < 0.001) were significantly associated with gender. CONCLUSIONS This work revealed the mutational landscape and characteristics of 30 core driver genes in a LUAD cohort. Co-mutated genes and genes associated with gender and age indicate their different roles in the corresponding subgroup of the LUAD.
Collapse
Affiliation(s)
- Jinmao Li
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Xianyun Li
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Hao Guo
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Shuni Zhou
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Hua Ding
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Bo Li
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Wei Li
- Wuhan Frasergen Bioinformatics Co., Ltd, Wuhan, China
| | - Bo Zhang
- Wuhan Frasergen Bioinformatics Co., Ltd, Wuhan, China
| | - Yi Zou
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China.
| |
Collapse
|
18
|
Ponchel T, Loeffler E, Ancel J, Brisebarre A, Lalun N, Dalstein V, Durlach A, Deslée G, Dedieu S, Polette M, Nawrocki-Raby B. LRP1 involvement in FHIT-regulated HER2 signaling in non-small cell lung cancer. Eur J Cell Biol 2025; 104:151475. [PMID: 39752787 DOI: 10.1016/j.ejcb.2024.151475] [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: 05/17/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025] Open
Abstract
The tumor suppressor fragile histidine triad (FHIT) is frequently lost in non-small cell lung cancer (NSCLC). We previously showed that a down-regulation of FHIT causes an up-regulation of the activity of HER2 associated to an epithelial-mesenchymal transition (EMT) and that lung tumor cells harboring a FHITlow/pHER2high phenotype are sensitive to anti-HER2 drugs. Here, we sought to decipher the FHIT-regulated HER2 signaling pathway in NSCLC. Transcriptomic analysis of tumor cells isolated from NSCLC revealed the endocytic receptor low density lipoprotein receptor-related protein 1 (LRP1), a central regulator of membrane trafficking and cell signaling, as a potential player of this signaling. In a cohort of 80 NSCLC assessed by immunohistochemistry, we found a significant association between a low FHIT expression and a high pHER2 and LRP1 expression by tumor cells. Experiments of FHIT silencing showed that FHIT regulated LRP1 expression both at the mRNA and protein levels in lung cell lines. Analyzing the relationship between LRP1 and HER2, we observed that an anti-HER2 targeted therapy reversed LRP1 overexpression induced by FHIT silencing whereas LRP1 silencing did not affect HER2 activity. Studying the functional role of LRP1, we showed that cell proliferation and invasion induced by FHIT silencing were LRP1-dependent. In addition, we found that the induction of vimentin upon FHIT inactivation was counteracted by LRP1 silencing. These results suggest that LRP1 acts downstream of HER2 to induce EMT and tumor progression following FHIT loss. Dual targeting of HER2 and LRP1 might represent a therapeutic strategy to more efficiently inhibit HER2 signaling in FHIT-negative NSCLC.
Collapse
Affiliation(s)
- Théophile Ponchel
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France
| | - Emma Loeffler
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France
| | - Julien Ancel
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France; CHU de Reims, Hôpital Maison Blanche, Service de Pneumologie, Reims, France
| | - Audrey Brisebarre
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France
| | - Nathalie Lalun
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France
| | - Véronique Dalstein
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France; CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, Reims, France
| | - Anne Durlach
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France; CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, Reims, France
| | - Gaëtan Deslée
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France; CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, Reims, France
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, UMR 7369, Reims, France
| | - Myriam Polette
- Université de Reims Champagne-Ardenne, INSERM, P3Cell, UMR-S 1250, Reims, France; CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, Reims, France
| | | |
Collapse
|
19
|
Yang X, Ma B, Liu Y, Zhou J, Guo J, Peng Y, Bai Y, Wu J, Hu D. SSBP1 positively regulates RRM2, affecting epithelial mesenchymal transition and cell cycle arrest in human lung adenocarcinoma cells. Cell Signal 2025; 127:111552. [PMID: 39643024 DOI: 10.1016/j.cellsig.2024.111552] [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: 10/15/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Progression of lung adenocarcinoma (LUAD) is frequently associated with alterations in epithelial-mesenchymal transition (EMT) and cell cycle. Our study analyzed the Cancer Genome Atlas (TCGA) database and identified a positive correlation between high expression of SSBP1 in LUAD tumor tissues and poor prognosis (p < 0.05), with an AUC of 0.853, suggesting that SSBP1 could serve as a prognostic biomarker. In vitro experiments, including siRNA-mediated SSBP1 knockdown and subsequent cell cloning and Transwell assays, revealed significant inhibition of proliferation, migration, and cell cycle progression in LUAD cells (p < 0.05). In vivo mouse model experiments further confirmed that SSBP1 knockdown inhibits tumor burden (p < 0.05). Mechanistic investigations, integrating pathway enrichment analysis with molecular biology techniques, identified RRM2 as a downstream target of SSBP1, and RRM2 knockdown similarly suppressed LUAD cell proliferation, migration, and cell cycle progression (p < 0.05). These findings indicate that SSBP1 promotes EMT and cell cycle progression in LUAD cells by positively regulating RRM2, thereby accelerating disease progression. Collectively, our study not only confirms the potential role of SSBP1 in LUAD but also provides a theoretical foundation for therapeutic strategies targeting the SSBP1/RRM2 axis, potentially offering new therapeutic targets for LUAD patients.
Collapse
Affiliation(s)
- Xingyu Yang
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Bingfeng Ma
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yafeng Liu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jiawei Zhou
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jianqiang Guo
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yuanyuan Peng
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Ying Bai
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Jing Wu
- Joint Research Center for Occupational Medicine and Health of IHM, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Dong Hu
- The First Affiliated Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), School of Medicine, Huainan City, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei City, China.
| |
Collapse
|
20
|
Uehara Y, Izumi H, Taki T, Sakai T, Udagawa H, Sugiyama E, Umemura S, Zenke Y, Matsumoto S, Yoh K, Kubota S, Aokage K, Sakamoto N, Sakashita S, Kojima M, Nagamine M, Hosomi Y, Tsuboi M, Goto K, Ishii G. Solid Predominant Histology and High Podoplanin Expression in Cancer-Associated Fibroblast Predict Primary Resistance to Osimertinib in EGFR-Mutated Lung Adenocarcinoma. JTO Clin Res Rep 2025; 6:100779. [PMID: 40007550 PMCID: PMC11850751 DOI: 10.1016/j.jtocrr.2024.100779] [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: 05/03/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 02/27/2025] Open
Abstract
Introduction Resistance to EGFR tyrosine kinase inhibitors is influenced by tumor-intrinsic and -extrinsic factors. We investigated the impact of tumor cell histology and tumor microenvironment on the efficacy of osimertinib. Methods We evaluated surgically resected adenocarcinoma from patients treated with first-line osimertinib at the National Cancer Center Hospital East (2016-2023), evaluating clinicopathologic characteristics, tumor cell histology, podoplanin expression in cancer-associated fibroblasts (CAFs) identified by immunohistochemistry, and outcomes. We also investigated HGF mRNA expression levels, using The Cancer Genome Atlas Program and Singapore Oncology Data Portal cohorts. Results The study included 93 patients. Solid (n = 19) versus non-solid predominant (n = 74) histology was not associated with worse disease-free survival after surgery (p = 0.12), but was significantly associated with worse progression-free survival (PFS) and overall survival following osimertinib treatment (p = 0.026, p = 0.004). Similarly, high-podoplanin (n = 31) versus low-podoplanin (n = 62) expression in CAFs was not associated with worse disease-free survival after surgery (p = 0.65), but was significantly associated with worse PFS and showed a trend towards worse overall survival following osimertinib treatment (p < 0.001, p = 0.11). In the multivariable analysis, solid predominant histology and high-podoplanin expression in CAFs were independently associated with worse PFS. In the cohorts of The Cancer Genome Atlas Program and Singapore Oncology Data Portal, EGFR-mutated lung adenocarcinoma with solid predominant histology or high-podoplanin expression exhibited significantly higher HGF expression. Conclusions Solid predominant histology and high-podoplanin expression in CAFs predicted osimertinib resistance, potentially guiding the selection of patients for more intensive treatments beyond osimertinib monotherapy.
Collapse
Affiliation(s)
- Yuji Uehara
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hiroki Izumi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuro Taki
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuya Sakai
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Eri Sugiyama
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Zenke
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shoko Kubota
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keiju Aokage
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Sakashita
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Michiko Nagamine
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Innovative Pathology and Laboratory Medicine, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
21
|
Shi R, Sun J, Zhou Z, Shi M, Wang X, Gao Z, Zhao T, Li M, Shu Y. Integration of multiple machine learning approaches develops a gene mutation-based classifier for accurate immunotherapy outcomes. NPJ Precis Oncol 2025; 9:54. [PMID: 40011681 DOI: 10.1038/s41698-025-00842-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
In addition to traditional biomarkers like PD-(L)1 expression and tumor mutation burden (TMB), more reliable methods for predicting immune checkpoint blockade (ICB) response in cancer patients are urgently needed. This study utilized multiple machine learning approaches on nonsynonymous mutations to identify key mutations that are most significantly correlated to ICB response. We proposed a classifier, Gene mutation-based Predictive Signature (GPS), to categorize patients based on their predicted response and clinical outcomes post-ICB therapy. GPS outperformed conventional predictors when validated in independent cohorts. Multi-omics analysis and multiplex immunohistochemistry (mIHC) revealed insights into tumor immunogenicity, immune responses, and the tumor microenvironment (TME) in lung adenocarcinoma (LUAD) across different GPS groups. Finally, we validated distinct responses of different GPS samples to ICB in an ex-vivo tumor organoid-PBMC co-culture model. Overall, our findings highlight a simple, robust classifier for accurate ICB response prediction, which could reduce costs, shorten testing times, and facilitate clinical implementation.
Collapse
Affiliation(s)
- Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Sun
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meiqi Shi
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xin Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Zhaojia Gao
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Tianyu Zhao
- Institute and Clinic for Occupational, Social and Environmental Medicine, LMU University Hospital Munich, Munich, Germany
| | - Minglun Li
- Department of Radiation Oncology, Lueneburg Municipal Hospital, Lueneburg, Germany
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
22
|
Xin S, Wen M, Tian Y, Dong H, Wan Z, Jiang S, Meng F, Xiong Y, Han Y. Impact of histopathological subtypes on invasive lung adenocarcinoma: from epidemiology to tumour microenvironment to therapeutic strategies. World J Surg Oncol 2025; 23:66. [PMID: 40016762 PMCID: PMC11866629 DOI: 10.1186/s12957-025-03701-9] [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: 09/04/2024] [Accepted: 02/02/2025] [Indexed: 03/01/2025] Open
Abstract
Lung adenocarcinoma is the most prevalent type of lung cancer, with invasive lung adenocarcinoma being the most common subtype. Screening and early treatment of high-risk individuals have improved survival; however, significant differences in prognosis still exist among patients at the same stage, especially in the early stages. Invasive lung adenocarcinoma has different histological morphologies and biological characteristics that can distinguish its prognosis. Notably, several studies have found that the pathological subtypes of invasive lung adenocarcinoma are closely associated with clinical treatment. This review summarised the distribution of various pathological subtypes of invasive lung adenocarcinoma in the population and their relationship with sex, smoking, imaging features, and other histological characteristics. We comprehensively analysed the genetic characteristics and biomarkers of the different pathological subtypes of invasive lung adenocarcinoma. Understanding the interaction between the pathological subtypes of invasive lung adenocarcinoma and the tumour microenvironment helps to reveal new therapeutic targets for lung adenocarcinoma. We also extensively reviewed the prognosis of various pathological subtypes and their effects on selecting surgical methods and adjuvant therapy and explored future treatment strategies.
Collapse
Affiliation(s)
- Shaowei Xin
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
- Department of Thoracic Surgery, 962 Hospital of the Joint Logistics Support Force, Harbin, China
| | - Miaomiao Wen
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yahui Tian
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Honghong Dong
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Zitong Wan
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- College of Life Sciences, Northwestern University, Xi'an, 710069, China
| | - Suxin Jiang
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Fancheng Meng
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
- Innovation Center for Advanced Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital and PLA Medical School, Beijing, China.
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Shaanxi, , Xi'an, 710038, China.
| | - Yong Han
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China.
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, 30 Fucheng Road, Haidian District, Shaanxi, , Beijing, 100142, China.
| |
Collapse
|
23
|
Piazzolla M, Parente P, Centra F, Fabrizio FP, Delcuratolo MD, Centonza A, Di Micco CM, Mastroianno M, Delli Muti F, Fiordelisi F, Ferretti G, Graziano P, Muscarella LA. Distribution of EGFR and KRAS Mutations in Patients with Surgically Resected Non-Small Cell Lung Cancer from Southern Italy: Real-Life Data from a Single Institution and Literature Review. Cancers (Basel) 2025; 17:730. [PMID: 40075579 PMCID: PMC11899559 DOI: 10.3390/cancers17050730] [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: 01/23/2025] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES The identification of driver mutations in NSCLC such as those in the EGFR and KRAS genes has revolutionized the understanding and management of many lung cancer patients and has opened up a new scenario in the early disease stages in terms of therapeutic options (EGFR) and prognosis (KRAS). Data on prevalence rates and disease stage distributions of EGFR and KRAS mutations in surgically resected NSCLC are growing, but in Southern Italy, estimation is limited, since upfront EGFR testing in early-stage adenocarcinoma has been only recently introduced according to the current guidelines in clinical practice, whereas KRAS screening is usually uninvestigated in resected NSCLC. In this real-life study of a single institution in the Apulia Region, we provide an overview of the epidemiological distribution of EGFR and KRAS mutations in patients in Southern Italy with resected NSCLC, highlighting their prevalence, clinical significance, and correlation with demographic and pathological factors. A literature review was also performed to compare our findings with the most recent available data from the screening of Italian cohorts of advanced and surgically resected NSCLC patients. METHODS Data from 149 patients coming from Southern Italy with surgically resected NSCLC were retrospectively collected over a period of 16 years. EGFR and KRAS mutation screenings were performed and correlated with clinical and pathological data. RESULTS In total, 24 out of 149 NSCLC (16%) patients harbored an EGFR mutation. Exon 19 deletions and missense p.L858R mutations of the EGFR gene have quite similar frequencies (46%) and were more observed in never smokers (p < 0.001) and female (p < 0.001) patients with the adenocarcinoma histotype. KRAS gene mutations were detected in 31.5% of cases, with missense p.G12C (32%), p.G12V (28%), and p.G12D (17%) mutations as the most frequent ones. Neither EGFR nor KRAS mutational status were found to impact overall survival (OS) in our study cohort. CONCLUSIONS Our findings improve the understanding of lung cancer genetics in a small and homogeneous area of Southern Italy and guide future research. The EGFR and KRAS mutations in NSCLC resected patients from Southern Italy showed a global similar incidence compared to other recently described Italian cohorts of advanced and early-stage NSCLC, with a higher frequency of exon19 EGFR deletions. No prognostic impact was observed for both EGFR and KRAS status, but additional investigations on a larger cohort are needed.
Collapse
Affiliation(s)
- Michele Piazzolla
- Unit of Thoracic Surgery, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.P.); (G.F.)
| | - Paola Parente
- Unit of Pathology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (P.P.); (F.F.); (P.G.)
| | - Flavia Centra
- Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy (F.P.F.); (F.D.M.)
| | - Federico Pio Fabrizio
- Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy (F.P.F.); (F.D.M.)
| | - Marco Donatello Delcuratolo
- Unit of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.D.D.); (C.M.D.M.)
| | - Antonella Centonza
- Unit of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.D.D.); (C.M.D.M.)
| | - Concetta Martina Di Micco
- Unit of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.D.D.); (C.M.D.M.)
| | - Mario Mastroianno
- Scientific Direction, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Francesco Delli Muti
- Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy (F.P.F.); (F.D.M.)
| | - Fabiola Fiordelisi
- Unit of Pathology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (P.P.); (F.F.); (P.G.)
| | - Gianmaria Ferretti
- Unit of Thoracic Surgery, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (M.P.); (G.F.)
| | - Paolo Graziano
- Unit of Pathology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (P.P.); (F.F.); (P.G.)
| | - Lucia Anna Muscarella
- Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy (F.P.F.); (F.D.M.)
| |
Collapse
|
24
|
Shen Y, Fu S, Liu X, Liu J, Fu Y, Zhao Y, Wang X, Jiang X, Zhang J. Gene Mutation Characteristics and Prognostic Significance in Acute Myeloid Leukemia Patients From Northeast China. Hum Mutat 2025; 2025:7730186. [PMID: 40226311 PMCID: PMC11918257 DOI: 10.1155/humu/7730186] [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: 04/25/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 04/15/2025]
Abstract
A great part of studies on the correlation between gene mutations and prognosis in acute myeloid leukemia (AML) patients are based on Western populations. To profile the genomic landscape of AML patients in Northeast China, we retrospectively analyzed the clinical data of 377 newly diagnosed AML patients in Shengjing Hospital of China Medical University from 2016 to 2022 and compared them with data from other populations with different genetic backgrounds. The mutation status of NPM1, FLT3-ITD, FLT3-TKD, CEBPA (CCAT enhancer binding protein alpha), ASXL1, TET2, KIT, DNMT3A (DNA methyltransferase 3A), IDH1, IDH2, EZH2 (enhancer of zeste 2), RUNX1, TP53, NRAS, and GATA2 was acquired by next-generation sequencing (NGS) technology; meanwhile, the clinical data of the patients were collected. The Cox regression model was used to analyze factors affecting patient survival and the impact of CEBPA and DNMT3A mutation on prognosis, and the results were different from those in other populations. Seventy-seven of 377 patients (20.4%) were detected with CEBPA mutations, which was higher than the 2%-6% in the Caucasian population. In the CEBPAdm patients who did not receive bone marrow transplantation, the prognosis of male patients (n = 18) was significantly better than that of female patients (n = 21) (p = 0.0242). Sixty-three of 377 patients (16.7%) carried the DNMT3A mutation, which was lower than the mutation frequency of 20.9% in the German-Austrian population, and the prognosis of these patients was significantly poorer (p = 0.0052). In addition, the prognostic evaluation value of the DNMT3A mutation in AML patients was not affected regardless of the presence of the NPM1 and FLT3-ITD comutation (p > 0.05), nor the mutation site of DNMT3A. In conclusion, for the Northeastern Chinese population, the prognosis of male patients with CEBPAdm was more favorable than that of female patients, and the DNMT3A mutation serves as an independent predictor of poor prognosis in AML. These results highlighted the central role of genetic background in precision medicine strategies and further emphasized the importance of the clinical characteristics of AML gene mutations in the Chinese population.
Collapse
Affiliation(s)
- Yiyang Shen
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuang Fu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuan Liu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianing Liu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Fu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yue Zhao
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinxin Wang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xujian Jiang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jihong Zhang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Zhang C, Yang C, Shi Q. Effects of Tp53 Gene Mutations on the Survival of Non-Small Cell Lung Cancer (NSCLC); A Short Review. Cancer Manag Res 2025; 17:65-82. [PMID: 39830995 PMCID: PMC11742633 DOI: 10.2147/cmar.s495006] [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: 09/06/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. Mutations within the TP53 gene represent critical molecular events in NSCLC, contributing to the tumorigenesis in the pulmonary epithelial tissues. TP53 is a widely researched prognostic indicator in NSCLC, and pathological investigations have revealed a weak to mild negative predictive effect for TP53. Mutated p53 protein may have some pro-oncogenic impact, and the variations may change tumor inhibitors into oncogenes. The diverse mutational spectrum of TP53 in NSCLC with different mutations is linked to varied treatment responses. In contrast, first-line chemotherapeutics to this progress are limited, however, randomized trials with new chemotherapeutics have shown significant survival benefits. This review highlighted the critical influence of TP53 gene mutations on pathological-sensitivity and overall survival outcomes in NSCLC. Further research is needed to explore TP53 mutation-specific pathways and their effects on NSCLC progression and treatment effectiveness.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Oncology, Anhui Chest Hospital, Hefei, 230022, People’s Republic of China
- Anhui Medical University Clinical College of Chest, Hefei, 230022, People’s Republic of China
| | - Chao Yang
- Department of Urology, Anhui Provincial Children’s Hospital, Hefei, 230022, People’s Republic of China
| | - Qingming Shi
- Department of Oncology, Anhui Chest Hospital, Hefei, 230022, People’s Republic of China
- Anhui Medical University Clinical College of Chest, Hefei, 230022, People’s Republic of China
| |
Collapse
|
26
|
Wang Z, Yuan X, Sun K, Wu F, Liu K, Jin Y, Chervova O, Nie Y, Yang A, Jin Y, Li J, Li Y, Yang F, Wang J, Beck S, Carbone D, Jiang G, Chen K. Optimizing the NGS-based discrimination of multiple lung cancers from the perspective of evolution. NPJ Precis Oncol 2025; 9:14. [PMID: 39809905 PMCID: PMC11733135 DOI: 10.1038/s41698-024-00786-5] [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: 04/01/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Next-generation sequencing (NGS) offers a promising approach for differentiating multiple primary lung cancers (MPLC) from intrapulmonary metastasis (IPM), though panel selection and clonal interpretation remain challenging. Whole-exome sequencing (WES) data from 80 lung cancer samples were utilized to simulate MPLC and IPM, with various sequenced panels constructed through gene subsampling. Two clonal interpretation approaches primarily applied in clinical practice, MoleA (based on shared mutation comparison) and MoleB (based on probability calculation), were subsequently evaluated. ROC analysis highlighted MoleB's superior performance, especially with the NCCNplus panel (AUC = 0.950 ± 0.002) and pancancer MoleA (AUC = 0.792 ± 0.004). In two independent cohorts (WES cohort, N = 42 and non-WES cohort, N = 94), NGS-based methodologies effectively stratified disease-free survival, with NCCNplus MoleB further predicting prognosis. Phylogenetic analysis further revealed evolutionary distinctions between MPLC and IPM, establishing an optimized NGS-based framework for differentiating multiple lung cancers.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
| | - Xiaoqiu Yuan
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Peking University Health Science Center, Beijing, China
| | - Kunkun Sun
- Department of Pathology, Peking University People's Hospital, Beijing, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, Hunan, China
- Changsha Thoracic Cancer Prevention and Treatment Technology Innovation Center, Changsha, Hunan, China
| | - Ke Liu
- Berry Oncology Corporation, Beijing, China
| | - Yiruo Jin
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Peking University Health Science Center, Beijing, China
| | - Olga Chervova
- University College London Cancer Institute, University College London, London, UK
| | - Yuntao Nie
- China-Japan Friendship Hospital, Beijing, China
| | | | - Yichen Jin
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
| | - Jing Li
- Berry Oncology Corporation, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
| | - Jun Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
| | - Stephan Beck
- University College London Cancer Institute, University College London, London, UK
| | - David Carbone
- James Thoracic Oncology Center, Ohio State University, Columbus, USA
| | - Guanchao Jiang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
| | - Kezhong Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, 100044, China.
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China.
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
27
|
Yang X, Li X, Huang K, Zhuang X. Evaluation of the efficacy of PD‑1/PD‑L1 inhibitor plus bevacizumab and chemotherapy for the treatment of patients with driver gene‑negative advanced‑stage lung adenocarcinoma: A retrospective cohort study. Oncol Lett 2025; 29:53. [PMID: 39584040 PMCID: PMC11582526 DOI: 10.3892/ol.2024.14799] [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: 02/22/2024] [Accepted: 05/10/2024] [Indexed: 11/26/2024] Open
Abstract
Driver gene-negative advanced-stage lung adenocarcinoma is associated with a poor prognosis and insufficient treatment options. The present study aimed to evaluate the efficacy and safety profile of a programmed cell death protein 1/programmed death-ligand 1 inhibitor plus bevacizumab and chemotherapy (PBC) regimen for the treatment of patients with driver gene-negative advanced-stage lung adenocarcinoma under real-world clinical conditions. Data from 65 patients with advanced-stage lung adenocarcinoma without sensitizing epidermal growth factor receptor, ALK receptor tyrosine kinase or ROS proto-oncogene 1 receptor tyrosine kinase mutations who received a PBC regimen or only a BC regimen were reviewed in the present retrospective cohort study. The results revealed that the objective response rate was higher (70.4 vs. 47.4%; P=0.065) in the PBC group compared with that in the BC group, while not reaching statistical significance. Progression-free survival (PFS) time was longer in the PBC group than in the BC group [median PFS: 10.8 months (95% confidence interval (CI), 7.2-14.4) vs. 7.6 months (95% CI, 5.0-10.2); P=0.016], while overall survival (OS) exhibited a non-significant trend to be longer in the PBC group compared with that in the BC group [median OS: 20.6 months (95% CI, 16.8-24.4) vs. 15.9 months (95% CI, 11.8-20.0); P=0.115]. Following adjustment by multivariate Cox analysis, the PBC (vs. BC) regimen was found to be independently associated with an improved PFS time (P=0.045). The common adverse events in the PBC group were neutropenia, alopecia, leukopenia, nausea and vomiting, fatigue, anemia and peripheral neuropathy. Moreover, the incidence of each adverse event did not differ significantly between the PBC and BC groups. In conclusion, the present study demonstrated that the PBC regimen serves as a superior treatment option for patients with driver gene-negative advanced-stage lung adenocarcinoma; however, further verification of its efficacy is still required.
Collapse
Affiliation(s)
- Xiaozun Yang
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610000, P.R. China
| | - Xin Li
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610000, P.R. China
| | - Ke Huang
- Department of Thoracic Surgery, Sichuan Jianzhu Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Xiang Zhuang
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
28
|
Wang J, Zhou W, Xu Y, Duan J, Zhou Q, Wang G, Li L, Xu C, Wang W, Cai S, Wang Z, Wang J. Antithetical impacts of deleterious LRP1B mutations in non-squamous and squamous NSCLCs on predicting benefits from immune checkpoint inhibitor alone or with chemotherapy over chemotherapy alone: retrospective analyses of the POPLAR/OAK and CHOICE-01 trials. SCIENCE CHINA. LIFE SCIENCES 2025; 68:249-262. [PMID: 39276256 DOI: 10.1007/s11427-023-2554-y] [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: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 09/16/2024]
Abstract
In non-small cell lung cancers, the non-squamous and squamous subtypes (nsqNSCLC and sqNSCLC) exhibit disparities in pathophysiology, tumor immunology, and potential genomic correlates affecting responses to immune checkpoint inhibitor (ICI)-based treatments. In our in-house training cohort (n=85), the presence of the LRP1B deleterious mutation (LRP1B-del) was associated with longer and shorter progression-free survival (PFS) on ICIs alone in nsqNSCLCs and sqNSCLCs, respectively (Pinteraction=0.008). These results were validated using a larger public ICI cohort (n=208, Pinteraction<0.001). Multiplex immunofluorescence staining revealed an association between LRP1B-del and increased and decreased numbers of tumor-infiltrating CD8+ T cells in nsqNSCLCs (P=0.040) and sqNSCLCs (P=0.014), respectively. In the POPLAR/OAK cohort, nsqNSCLCs with LRP1B-del demonstrated improved PFS benefits from atezolizumab over docetaxel (hazard ratio (HR) =0.70, P=0.046), whereas this benefit was negligible in those without LRP1B-del (HR=1.05, P=0.64). Conversely, sqNSCLCs without LRP1B-del benefited more from atezolizumab (HR=0.60, P=0.002) than those with LRP1B-del (HR=1.30, P=0.31). Consistent results were observed in the in-house CHOICE-01 cohort, in which nsqNSCLCs with LRP1B-del and sqNSCLCs without LRP1B-del benefited more from toripalimab plus chemotherapy than from chemotherapy alone (Pinteraction=0.008). This multi-cohort study delineates the antithetical impacts of LRP1B-del in nsqNSCLCs and sqNSCLCs on predicting the benefits from ICI alone or with chemotherapy over chemotherapy alone. Our findings highlight the distinct clinical utility of LRP1B-del in guiding treatment choices for nsqNSCLCs and sqNSCLCs, emphasizing the necessity for a detailed analysis based on pathological subtypes when investigating biomarkers for cancer therapeutics.
Collapse
Affiliation(s)
- Jinliang Wang
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Wenyong Zhou
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yu Xu
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Jianchun Duan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | | | | | - Leo Li
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Chunwei Xu
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310000, China
| | - Wenxian Wang
- Department of Clinical Trial, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310022, China
| | - Shangli Cai
- Burning Rock Biotech, Guangzhou, 510300, China.
| | - Zhijie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
29
|
Wang J, Guo C, Wang J, Zhang X, Qi J, Huang X, Hu Z, Wang H, Hong B. Tumor Mutation Signature Reveals the Risk Factors of Lung Adenocarcinoma with EGFR or KRAS Mutation. Cancer Control 2025; 32:10732748241307363. [PMID: 39760242 DOI: 10.1177/10732748241307363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
INTRODUCTION EGFR and KRAS mutations are frequently detected in lung adenocarcinoma (LUAD). Tumor mutational signature (TMS) determination is an approach to identify somatic mutational patterns associated with pathogenic factors. In this study, through the analysis of TMS, the underlying pathogenic factors of LUAD with EGFR and KRAS mutations were traced. METHODS This was a retrospective study. TMS of LUAD with KRAS and EGFR mutations from the TCGA, OncoSG, and MSK datasets was determined by two bioinformatics tools, namely the "MutationalPatterns" and "FitMS" packages. Elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) of LUAD clinical specimens was analyzed using capillary electrophoresis. RESULTS In LUAD with KRAS mutations, TMS analysis indicated that the smoking-related SBS4 signature was enriched. For LUAD with EGFR L858R mutation, the smoking-related SBS4 signature was enriched in the Western population from the TCGA database; however, the smoking-related SBS4 signature was not obvious in Asian LUAD patients. LUAD with EGFR exon19 deletion (19Del) exhibited stronger SBS15 signature, which was related to defective DNA mismatch repair. Capillary electrophoresis analysis showed that an EMAST locus was frequently instable in LUAD with EGFR 19Del. Different from the Western population, Asian LUAD patients with EGFR mutations exhibited the enrichment of SBS1, SBS2, and SBS13 signatures, which were associated with the endogenous mutation process of cytidine deamination. CONCLUSIONS TMS analysis reveals that smoking is associated with LUAD with KRAS mutations. Defective DNA mismatch repair and endogenous cytidine deamination are associated with LUAD with EGFR mutations, especially for the EGFR 19Del. The endogenous mutational process is stronger in Asian LUAD patients than Western LUAD patients.
Collapse
Affiliation(s)
- Jialiang Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Chang Guo
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Jiexiao Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Xiaopeng Zhang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Jian Qi
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Xiang Huang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Zongtao Hu
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Hongzhi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Bo Hong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| |
Collapse
|
30
|
Li Q, Zhang Y, Luo S, Zhang Z, Oberg AL, Kozono DE, Lu H, Sarkaria JN, Ma L, Wang L. Identify Non-mutational p53 Functional Deficiency in Human Cancers. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae064. [PMID: 39325855 PMCID: PMC11702981 DOI: 10.1093/gpbjnl/qzae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 07/23/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024]
Abstract
An accurate assessment of p53's functional statuses is critical for cancer genomic medicine. However, there is a significant challenge in identifying tumors with non-mutational p53 inactivation which is not detectable through DNA sequencing. These undetected cases are often misclassified as p53-normal, leading to inaccurate prognosis and downstream association analyses. To address this issue, we built the support vector machine (SVM) models to systematically reassess p53's functional statuses in TP53 wild-type (TP53WT) tumors from multiple The Cancer Genome Atlas (TCGA) cohorts. Cross-validation demonstrated the good performance of the SVM models with a mean area under the receiver operating characteristic curve (AUROC) of 0.9822, precision of 0.9747, and recall of 0.9784. Our study revealed that a significant proportion (87%-99%) of TP53WT tumors actually had compromised p53 function. Additional analyses uncovered that these genetically intact but functionally impaired (termed as predictively reduced function of p53 or TP53WT-pRF) tumors exhibited genomic and pathophysiologic features akin to TP53-mutant tumors: heightened genomic instability and elevated levels of hypoxia. Clinically, patients with TP53WT-pRF tumors experienced significantly shortened overall survival or progression-free survival compared to those with predictively normal function of p53 (TP53WT-pN) tumors, and these patients also displayed increased sensitivity to platinum-based chemotherapy and radiation therapy.
Collapse
Affiliation(s)
- Qianpeng Li
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sicheng Luo
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ann L Oberg
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - David E Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Hua Lu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lina Ma
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Rochester, Rochester, MN 55904, USA
| |
Collapse
|
31
|
Chen Y, Zhu X, Wang J, Hu J, Zhang J, Zhang X, Han L, Yu H, Hu H, Fei K, Zhang P, Zhang L. MAZ promotes tumor proliferation and immune evasion in lung adenocarcinoma. Oncogene 2024; 43:3619-3632. [PMID: 39424990 DOI: 10.1038/s41388-024-03194-y] [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: 05/21/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
Lung adenocarcinoma (LUAD) is the most dominant histological subtype of lung cancer and one of the most lethal malignancies. The identification of novel therapeutic targets is required for the treatment of LUAD. Here, we showed that MYC-associated zinc-finger protein (MAZ) is upregulated in LUAD tissues. MAZ expression levels are inversely correlated with patient survival. Silencing of MAZ decreased tumor proliferation and the expression of pro-tumorigenic chemokines and Galectin-9 (Gal-9), an immune checkpoint molecule. The pro-tumorigenic chemokines and Gal-9 induce immune suppression by recruitment of myeloid cells and inhibition of T cell activation, respectively. Mechanistically, MAZ transcriptionally regulates KRAS expression and activates its downstream AKT-NF-κB signaling pathway, which is crucial for tumor progression and immune evasion. Additionally, in vivo animal models and bioinformatic analyses indicated that MAZ suppression could enhance the efficacy of immune checkpoint blockade (ICB) therapy for LUAD. Overall, our results suggest that MAZ plays an important role in regulating cell proliferation and immune evasion via KRAS/AKT/NF-κB signaling in LUAD. Our findings offer a candidate molecular target for LUAD therapy, with implications for improving the efficacy of ICB therapy.
Collapse
Affiliation(s)
- Yan Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Xinsheng Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Jue Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Junjie Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Xun Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lu Han
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Huansha Yu
- Experimental Animal Center, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Haiyang Hu
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Ke Fei
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Lele Zhang
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
32
|
Luo G, Kumar H, Aldridge K, Rieger S, Han E, Jiang E, Chan ER, Soliman A, Mahdi H, Letterio JJ. A Core NRF2 Gene Set Defined Through Comprehensive Transcriptomic Analysis Predicts Selective Drug Resistance and Poor Multicancer Prognosis. Antioxid Redox Signal 2024; 41:1031-1050. [PMID: 39028025 DOI: 10.1089/ars.2023.0409] [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] [Indexed: 07/20/2024]
Abstract
Aims: The nuclear factor erythroid 2-related factor 2-Kelch-like ECH-associated protein 1 (NRF2-KEAP1) pathway plays an important role in the cellular response to oxidative stress but may also contribute to metabolic changes and drug resistance in cancer. However, despite its pervasiveness and important role, most of nuclear factor erythroid 2-related factor 2 (NRF2) target genes are defined in context-specific experiments and analysis, making it difficult to translate from one situation to another. Our study investigates whether a core NRF2 gene signature can be derived and used to represent NRF2 activation in various contexts, allowing better reproducibility and understanding of NRF2. Results: We define a core set of 14 upregulated NRF2 target genes from 7 RNA-sequencing datasets that we generated and analyzed. This NRF2 gene signature was validated using analyses of published datasets and gene sets. An NRF2 activity score based on expression of these core target genes correlates with resistance to drugs such as PX-12 and necrosulfonamide but not to paclitaxel or bardoxolone methyl. We validated these findings in our Kelch-like ECH-associated protein 1 (KEAP1) knockout cancer cell lines. Finally, our NRF2 score is prognostic for cancer survival and validated in additional independent cohorts for lung adenocarcinoma and also novel cancer types not associated with NRF2-KEAP1 mutations such as clear cell renal carcinoma, hepatocellular carcinoma, and acute myeloid leukemia. Innovation and Conclusions: These analyses define a core NRF2 gene signature that is robust, versatile, and useful for evaluating NRF2 activity and for predicting drug resistance and cancer prognosis. Using this gene signature, we uncovered novel selective drug resistance and cancer prognosis associated with NRF2 activation. Antioxid. Redox Signal. 41, 1031-1050.
Collapse
Affiliation(s)
- George Luo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Harshita Kumar
- Department of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Stevie Rieger
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - EunHyang Han
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Ethan Jiang
- Booth School of Business, University of Chicago, Chicago, Illinois, USA
| | - Ernest R Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ahmed Soliman
- Department of Pediatrics, SUNY Downstate Hospital, Brooklyn, New York, USA
| | - Haider Mahdi
- Magee Women's Research Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Magee Women's Hospital, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John J Letterio
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, Ohio, USA
- The Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
33
|
Wang H, Tang S, Wu Q, He Y, Zhu W, Xie X, Qin Z, Wang X, Zhou S, Yao S, Xu X, Guo C, Tong X, Han S, Chou YH, Wang Y, Wong KK, Yang CG, Chen L, Hu L, Ji H. Integrative study of lung cancer adeno-to-squamous transition in EGFR TKI resistance identifies RAPGEF3 as a therapeutic target. Natl Sci Rev 2024; 11:nwae392. [PMID: 39687207 PMCID: PMC11647589 DOI: 10.1093/nsr/nwae392] [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: 01/02/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 12/18/2024] Open
Abstract
Although adeno-to-squamous transition (AST) has been observed in association with resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) in clinic, its causality, molecular mechanism and overcoming strategies remain largely unclear. We here demonstrate that squamous transition occurs concomitantly with TKI resistance in PC9-derived xenograft tumors. Perturbation of squamous transition via DNp63 overexpression or knockdown leads to significant changes in TKI responses, indicative of a direct causal link between squamous transition and TKI resistance. Integrative RNA-seq, ATAC-seq analyses and functional studies reveal that FOXA1 plays an important role in maintaining adenomatous lineage and contributes to TKI sensitivity. FOXM1 overexpression together with FOXA1 knockout fully recapitulates squamous transition and TKI resistance in both PC9 xenografts and patient-derived xenograft (PDX) models. Importantly, pharmacological inhibition of RAPGEF3 combined with EGFR TKI efficiently overcomes TKI resistance, especially in RAPGEF3high PDXs. Our findings provide novel mechanistic insights into squamous transition and therapeutic strategy to overcome EGFR TKI resistance in lung cancer.
Collapse
Affiliation(s)
- Hua Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shijie Tang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qibiao Wu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200092, China
| | - Weikang Zhu
- Center for Excellence in Mathematical Sciences, National Center for Mathematics and Interdisciplinary Sciences, Key Laboratory of Management, Decision and Information System, Hua Loo-Keng Center for Mathematical Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
| | - Xinyun Xie
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhen Qin
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xue Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiyu Zhou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shun Yao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoling Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Chenchen Guo
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyuan Tong
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuo Han
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yueh-Hung Chou
- Department of Anatomical Pathology, Far Eastern Memorial Hospital, New Taipei City, Taiwan, China
| | - Yong Wang
- Center for Excellence in Mathematical Sciences, National Center for Mathematics and Interdisciplinary Sciences, Key Laboratory of Management, Decision and Information System, Hua Loo-Keng Center for Mathematical Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, NY 10016, USA
| | - Cai-Guang Yang
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Luonan Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Liang Hu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongbin Ji
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
34
|
Jha P, Joshi A, Mishra R, Biswal RP, Kulkarni PM, Limaye S, Babu G, Batra U, Malik P, Kumar R, Shah M, Menon N, Rauthan A, Kuriakose M, Ramachandran V, Noronha V, Kumar P, Prabhash K. Landscape of Clinically Relevant Genomic Alterations in the Indian Non-small Cell Lung Cancer Patients. Clin Lung Cancer 2024; 25:e420-e430.e20. [PMID: 39129089 DOI: 10.1016/j.cllc.2024.07.011] [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: 05/28/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND The genomic landscape of non-small cell lung cancer (NSCLC) in the Indian patients remains underexplored. We revealed distinctive genomic alterations of Indian NSCLC patients, thereby providing vital molecular insights for implementation of precision therapies. METHODS We analyzed the genomic profiles of 325 lung adenocarcinoma and 81 lung squamous carcinoma samples from Indian patients using targeted sequencing of 50 cancer related genes. Correlations between genomic alterations and clinical characteristics were computed using statistical analyses. Additionally, we identified distinct features of Indian NSCLC genomes by comparison across different ethnicities. RESULTS Our genomic analysis revealed several noticeable features of Indian NSCLC patients. Alterations in EGFR (45.8%), TP53 (27.4%), ALK (11.4%) and KRAS (10.2%) were predominant in adenocarcinoma, with 68% eligible for targeted therapies. Squamous carcinoma exhibited prevalent alterations in TP53 (40.7%), PIK3CA (17.3%), and CDKN2A (8.6%). We observed higher frequency of EGFR alterations (18.5%) in lung squamous carcinoma patients, significantly distinct from other ethnicities reported till date. Beyond established correlations, we observed 60% of PD-L1 negative squamous patients harbored TP53 alterations, suggesting intriguing therapeutic implications. CONCLUSIONS Our data revealed unique genomic variations of adenocarcinoma and squamous carcinoma patients, with significant indications for precision medicine and clinical practice of lung cancers. The study emphasizes the importance of clinical utility of NGS for routine diagnostics.
Collapse
Affiliation(s)
- Prerana Jha
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Asim Joshi
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Rohit Mishra
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Ranendra Pratap Biswal
- Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Pooja Mahesh Kulkarni
- Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Sewanti Limaye
- Department of Medical and Precision Oncology, Sir HN Reliance Foundation Hospital and Research Centre, Mumbai, Maharashtra, India
| | - Govind Babu
- Department of Medical Oncology, HCG Cancer Hospital, Bengaluru, India
| | - Ullas Batra
- Department of Medical Oncology, Section of Molecular Diagnostics, Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Prabhat Malik
- Department of Medical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Rajiv Kumar
- Department of Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Minit Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Rauthan
- Department of Medical Oncology, Manipal Hospital, Bengaluru, Karnataka, India
| | - Moni Kuriakose
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Venkataramanan Ramachandran
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Prashant Kumar
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India; Centre of Excellence for Cancer - Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh India.
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| |
Collapse
|
35
|
Kozono D, Hua X, Wu MC, Tolba KA, Waqar SN, Dragnev KH, Cheng H, Hirsch FR, Mack PC, Gray JE, Kelly K, Borghaei H, Herbst RS, Gandara DR, Redman MW. Lung-MAP Next-Generation Sequencing Analysis of Advanced Squamous Cell Lung Cancers (SWOG S1400). J Thorac Oncol 2024; 19:1618-1629. [PMID: 39111731 PMCID: PMC11625613 DOI: 10.1016/j.jtho.2024.07.024] [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: 03/13/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Squamous cell cancer (SqCC) is a lung cancer subtype with few targeted therapy options. Molecular characterization, that is, by next-generation sequencing (NGS), is needed to identify potential targets. Lung Cancer Master Protocol Southwest Oncology Group S1400 enrolled patients with previously treated stage IV or recurrent SqCC to assess NGS biomarkers for therapeutic sub-studies. METHODS Tumors underwent NGS using Foundation Medicine's FoundationOne research platform, which sequenced the exons and/or introns of 313 cancer-related genes. Mutually exclusive gene set analysis and Selected Events Linked by Evolutionary Conditions across Human Tumors were performed to identify mutually exclusive and co-occurring gene alterations. Comparisons were performed with data on 495 lung SqCC downloaded from The Cancer Genome Atlas. Cox proportional hazards models were used to assess associations between genetic variants and survival. RESULTS NGS data are reported for 1672 patients enrolled on S1400 between 2014 and 2019. Mutually exclusive gene set analysis identified two non-overlapping sets of mutually exclusive alterations with a false discovery rate of less than 15%: NFE2L2, KEAP1, and PARP4; and CDKN2A and RB1. PARP4, a relatively uncharacterized gene, showed three frequent mutations suggesting functional significance: 3116T>C (I1039T), 3176A>G (Q1059R), and 3509C>T (T1170I). When taken together, NFE2L2 and KEAP1 alterations were associated with poorer survival. CONCLUSIONS As the largest dataset to date of lung SqCC profiled on a clinical trial, the S1400 NGS dataset establishes a rich resource for biomarker discovery. Mutual exclusivity of PARP4 and NFE2L2 or KEAP1 alterations suggests that PARP4 may have an uncharacterized role in a key pathway known to impact oxidative stress response and treatment resistance.
Collapse
Affiliation(s)
- David Kozono
- Department of Radiation Oncology, Dana-Farber Brigham Cancer Center, Boston, Massachusetts.
| | - Xing Hua
- SWOG Statistics and Data Management Center, Seattle, Washington; Biostatistics Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Michael C Wu
- SWOG Statistics and Data Management Center, Seattle, Washington; Biostatistics Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Saiama N Waqar
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Haiying Cheng
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Fred R Hirsch
- Mt. Sinai Health System Center for Thoracic Oncology, Tisch Cancer Institute, New York, New York
| | - Philip C Mack
- Mt. Sinai Health System Center for Thoracic Oncology, Tisch Cancer Institute, New York, New York
| | - Jhanelle E Gray
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Karen Kelly
- Division of Hematology and Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California
| | - Hossein Borghaei
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Roy S Herbst
- Section of Medical Oncology, Yale University, New Haven, Connecticut
| | - David R Gandara
- Division of Hematology and Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California
| | - Mary W Redman
- SWOG Statistics and Data Management Center, Seattle, Washington; Biostatistics Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
36
|
Li H, Liu J, Zhang L, Xu Y, Wang X, Lan S, Cui P, Wang G, Cai S, Cheng Y. Mutation-guided chemotherapy-free strategy in first-line immunotherapy for low PD-L1-expressing non-squamous NSCLC. J Immunother Cancer 2024; 12:e009693. [PMID: 39615893 PMCID: PMC11624766 DOI: 10.1136/jitc-2024-009693] [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: 05/22/2024] [Accepted: 11/05/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The necessity of platinum-doublet chemotherapy in first-line immunotherapy for non-squamous non-small cell lung cancer (nsqNSCLC) with programmed death-ligand 1 (PD-L1) expression on less than 50% of tumor cells remains poorly investigated. Biomarkers predicting this necessity can guide chemotherapy-free treatment to minimize unnecessary toxicity. METHODS Treated with immune checkpoint inhibitor monotherapy (ICI-mono), chemotherapy, or combination (ICI-chemo), 790 low PD-L1-expressing nsqNSCLCs (in-house: n=83; public: n=707) were analyzed for development and validation of the interaction score for additional chemotherapy (ISAC). Transcriptomic (public, n=11) and multiplex immunofluorescence data (in-house, n=100) were analyzed to evaluate the immune microenvironment. RESULTS ICI-chemo, compared with ICI-mono, tended to prolong progression-free survival (PFS; HR=0.72, p=0.004) and overall survival (OS; HR=0.77, p=0.071) as first-line therapy in low PD-L1-expressing nsqNSCLCs. The added value of chemotherapy was observed in the ISAC-low subgroup (PFS: HR=0.48, p<0.001; OS: HR=0.53, p=0.001) rather than the ISAC-high subgroup (PFS: HR=1.08, p=0.65; OS: HR=1.14, p=0.56). This predictive utility was independent of tumor mutational burden and PD-L1 expression, indicated by subgroup and multivariable analyses. A high ISAC was associated with adaptive immune resistance reflected by more proinflammatory (eg, CD8+ T cells and M1 macrophages) rather than anti-inflammatory tumor-infiltrating immune cells (eg, M2 macrophages) and high expression of immune checkpoints except for PD-L1 (eg, programmed cell death protein-1). CONCLUSION A high ISAC was identified as a significant predictor for virtually no added value of platinum-doublet chemotherapy for first-line ICI treatment in low PD-L1-expressing nsqNSCLC. Our findings may help refine personalized therapeutic strategies for nsqNSCLC, thereby improving efficacy and reducing undue toxicity.
Collapse
Affiliation(s)
- Hui Li
- Translational Oncology Research Lab, Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, Jilin, China
| | - Jingjing Liu
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, Jilin, China
| | - Liang Zhang
- Oncology Department, Jilin Cancer Hospital, Changchun, Jilin, China
| | - Yu Xu
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Xinyue Wang
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, Jilin, China
| | - Shaowei Lan
- Translational Oncology Research Lab, Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, Jilin, China
| | - Peng Cui
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | | | - Shangli Cai
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, Jilin, China
| |
Collapse
|
37
|
Zhao Y, Zhang D, Meng B, Zhang Y, Ma S, Zeng J, Wang X, Peng T, Gong X, Zhai R, Dong L, Jiang Y, Dai X, Fang X, Jia W. Integrated proteomic and glycoproteomic analysis reveals heterogeneity and molecular signatures of brain metastases from lung adenocarcinomas. Cancer Lett 2024; 605:217262. [PMID: 39341452 DOI: 10.1016/j.canlet.2024.217262] [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: 05/09/2024] [Revised: 07/26/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024]
Abstract
Brain metastasis is a major cause of poor prognosis and death in lung adenocarcinoma (LUAD); however, the understanding of therapeutic strategies and mechanisms for brain metastases from LUAD (BM-LUAD) remains notably limited, especially at the proteomics levels. To address this issue, we conducted integrated proteomic and glycoproteomic analyses on 49 BM-LUAD tumors, revealing two distinct subtypes of the disease: BM-S1 and BM-S2. Whole exome sequencing analysis revealed that somatic mutations in STK11 and KEAP1, as well as copy number deletions on chr19p13.3, such as STK11, UQCR11, and SLC25A23, were more frequently detected in BM-S2. In BM-S1 tumors, we observed significant infiltration of GFAP + astrocytes, as evidenced by elevated levels of GFAP, GABRA2, GABRG1 and GAP43 proteins and an enrichment of astrocytic signatures in both our proteomic data and external spatial transcriptomic data. Conversely, BM-S2 tumors demonstrated higher levels of PD-1 immune cell infiltration, supported by the upregulation of PD-1 and LAG-3 genes. These findings suggest distinct microenvironmental adaptations required by the different BM-LUAD subtypes. Additionally, we observed unique glycosylation patterns between the subtypes, with increased fucosylation in BM-S1 and enhanced sialylation in BM-S2, primarily affected by glycosylation enzymes such as FUT9, B4GALT1, and ST6GAL1. Specifically, in BM-S2, these sialylation modifications are predominantly localized to the lysosomes, underscoring the critical role of N-glycosylation in the tumor progression of BM-LUAD. Overall, our study not only provides a comprehensive multi-omic data resource but also offers valuable biological insights into BM-LUAD, highlighting potential mechanisms and therapeutic targets for further investigation.
Collapse
Affiliation(s)
- Yang Zhao
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Bo Meng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Yong Zhang
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Jiaming Zeng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Tao Peng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Rui Zhai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Lianhua Dong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - You Jiang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xinhua Dai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China.
| | - Xiang Fang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China.
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
38
|
Zhang Y, Wang Y, Qian H. Multi-omics characterization and machine learning of lung adenocarcinoma molecular subtypes to guide precise chemotherapy and immunotherapy. Front Immunol 2024; 15:1497300. [PMID: 39669580 PMCID: PMC11634853 DOI: 10.3389/fimmu.2024.1497300] [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: 09/16/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a heterogeneous tumor characterized by diverse genetic and molecular alterations. Developing a multi-omics-based classification system for LUAD is urgently needed to advance biological understanding. Methods Data on clinical and pathological characteristics, genetic alterations, DNA methylation patterns, and the expression of mRNA, lncRNA, and microRNA, along with somatic mutations in LUAD patients, were gathered from the TCGA and GEO datasets. A computational workflow was utilized to merge multi-omics data from LUAD patients through 10 clustering techniques, which were paired with 10 machine learning methods to pinpoint detailed molecular subgroups and refine a prognostic risk model. The disparities in somatic mutations, copy number alterations, and immune cell infiltration between high- and low-risk groups were assessed. The effectiveness of immunotherapy in patients was evaluated through the TIDE and SubMap algorithms, supplemented by data from various immunotherapy groups. Furthermore, the Cancer Therapeutics Response Portal (CTRP) and the PRISM Repurposing dataset (PRISM) were employed to investigate new drug treatment approaches for LUAD. In the end, the role of SLC2A1 in tumor dynamics was examined using RT-PCR, immunohistochemistry, CCK-8, wound healing, and transwell tests. Results By employing multi-omics clustering, we discovered two unique cancer subtypes (CSs) linked to prognosis, with CS2 demonstrating a better outcome. A strong model made up of 17 genes was created using a random survival forest (RSF) method, which turned out to be an independent predictor of overall survival and showed reliable and impressive performance. The low-risk group not only had a better prognosis but also was more likely to display the "cold tumor" phenotype. On the other hand, individuals in the high-risk group showed a worse outlook and were more likely to respond positively to immunotherapy and six particular chemotherapy medications. Laboratory cell tests demonstrated that SLC2A1 is abundantly present in LUAD tissues and cells, greatly enhancing the proliferation and movement of LUAD cells. Conclusions Thorough examination of multi-omics data offers vital understanding and improves the molecular categorization of LUAD. Utilizing a powerful machine learning system, we highlight the immense potential of the riskscore in providing individualized risk evaluations and customized treatment suggestions for LUAD patients.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Laboratory Medicine, Guang’an People’s Hospital, Guang’an, Sichuan, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yuzhi Wang
- Department of Laboratory Medicine, Deyang People’s Hospital, Deyang, Sichuan, China
- Pathogenic Microbiology and Clinical Immunology Key Laboratory of Deyang City, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Haitao Qian
- Department of Anesthesiology, The First People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| |
Collapse
|
39
|
Xiong X, Liu Y, Pu D, Yang Z, Bi Z, Tian L, Li X. DeSide: A unified deep learning approach for cellular deconvolution of tumor microenvironment. Proc Natl Acad Sci U S A 2024; 121:e2407096121. [PMID: 39514318 PMCID: PMC11573681 DOI: 10.1073/pnas.2407096121] [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: 04/09/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Cellular deconvolution via bulk RNA sequencing (RNA-seq) presents a cost-effective and efficient alternative to experimental methods such as flow cytometry and single-cell RNA-seq (scRNA-seq) for analyzing the complex cellular composition of tumor microenvironments. Despite challenges due to heterogeneity within and among tumors, our innovative deep learning-based approach, DeSide, shows exceptional accuracy in estimating the proportions of 16 distinct cell types and subtypes within solid tumors. DeSide integrates biological pathways and assesses noncancerous cell types first, effectively sidestepping the issue of highly variable gene expression profiles (GEPs) associated with cancer cells. By leveraging scRNA-seq data from six cancer types and 185 cancer cell lines across 22 cancer types as references, our method introduces distinctive sampling and filtering techniques to generate a high-quality training set that closely replicates real tumor GEPs, based on The Cancer Genome Atlas (TCGA) bulk RNA-seq data. With this model and high-quality training set, DeSide outperforms existing methods in estimating tumor purity and the proportions of noncancerous cells within solid tumors. Our model precisely predicts cellular compositions across 19 cancer types from TCGA and proves its effectiveness with multiple additional external datasets. Crucially, DeSide enables the identification and analysis of combinatorial cell type pairs, facilitating the stratification of cancer patients into prognostically significant groups. This approach not only provides deeper insights into the dynamics of tumor biology but also highlights potential therapeutic targets by underscoring the importance of specific cell type or subtype interactions.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Physics, Hong Kong Baptist University, Hong Kong, China
| | - Yerong Liu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dandan Pu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Zedong Bi
- Lingang Laboratory, Shanghai 200031, China
| | - Liang Tian
- Department of Physics, Hong Kong Baptist University, Hong Kong, China
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
- Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong, China
- Institute of Systems Medicine and Health Sciences, Hong Kong Baptist University, Hong Kong, China
| | - Xuefei Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
40
|
Zhu B, McHale SS, Van Scoyk M, Riddick G, Wu PY, Chou CF, Chen CY, Winn RA. Gene expression-based modeling of overall survival in Black or African American patients with lung adenocarcinoma. Front Immunol 2024; 15:1478491. [PMID: 39588372 PMCID: PMC11586367 DOI: 10.3389/fimmu.2024.1478491] [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: 08/09/2024] [Accepted: 10/16/2024] [Indexed: 11/27/2024] Open
Abstract
Introduction Lung cancer is a leading cause of cancer-related deaths worldwide. Black/African American (B/AA) populations, in particular, exhibit the highest incidence and mortality rates of lung adenocarcinoma (LUAD) in the United States. Methods This study aims to explore gene expression patterns linked to LUAD in B/AA and case-matched white patients, with the goal of developing predictive models for prognosis. Leveraging RNA sequencing data from The Cancer Genome Atlas (TCGA) database, genes and pathways associated with overall survival (OS) were identified. Results The OS-associated genes in B/AA patients were distinct from those in white patients, showing predominant enrichment in immune-related pathways. Furthermore, mRNA co-expression network analysis revealed that OS-associated genes in B/AA patients had higher levels of interaction with various pathways, including those related to immunity, cell-ECM interaction, and specific intracellular signaling pathways. Notably, a potential B/AA-specific biomarker, C9orf64, demonstrated significant correlations with genes involved in immune response. Unsupervised machine learning algorithms stratified B/AA patients into groups with distinct survival outcomes, while supervised algorithms demonstrated a higher accuracy in predicting survival for B/AA LUAD patients compared to white patients. Discussion In total, this study explored OS-associated genes and pathways specific for B/AA LUAD patients. Further validation and clinical application of these findings are warranted to address disparities and improve outcomes in diverse patient populations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robert A. Winn
- Massey Comprehensive Cancer Center, Virginia Commonwealth University,
Richmond, VA, United States
| |
Collapse
|
41
|
Jiang Z, Gu Z, Yu X, Cheng T, Liu B. Research progress on the role of bypass activation mechanisms in resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Front Oncol 2024; 14:1447678. [PMID: 39582541 PMCID: PMC11581962 DOI: 10.3389/fonc.2024.1447678] [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: 06/12/2024] [Accepted: 09/25/2024] [Indexed: 11/26/2024] Open
Abstract
The clinical application of small molecule tyrosine kinase inhibitors (TKIs) has significantly improved the quality of life and prognosis of patients with non-small cell lung cancer (NSCLC) carrying driver genes. However, resistance to TKI treatment is inevitable. Bypass signal activation is one of the important reasons for TKI resistance. Although TKI drugs inhibit downstream signaling pathways of driver genes, key signaling pathways within tumor cells can still be persistently activated through bypass routes such as MET gene amplification, EGFR gene amplification, and AXL activation. This continuous activation maintains tumor cell growth and proliferation, leading to TKI resistance. The fundamental strategy to treat TKI resistance mediated by bypass activation involves simultaneously inhibiting the activated bypass signals and the original driver gene signaling pathways. Some clinical trials based on this combined treatment approach have yielded promising preliminary results, offering more treatment options for NSCLC patients with TKI resistance. Additionally, early identification of resistance mechanisms through liquid biopsy, personalized targeted therapy against these mechanisms, and preemptive targeting of drug-tolerant persistent cells may provide NSCLC patients with more sustained and effective treatment.
Collapse
Affiliation(s)
- Ziyang Jiang
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihan Gu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomin Yu
- Department of Emergency Medicine, West China Hospital, Sichuan University, West China School of Nursing, Sichuan University, Chengdu, China
- Institute of Disaster Medicine, Sichuan University, Chengdu, China
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Chengdu, China
| | - Tao Cheng
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bofu Liu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Zhang C, Wu Q, Yang H, Zhang H, Liu C, Yang B, Hu Q. Ferroptosis-related gene signature for predicting prognosis and identifying potential therapeutic drug in EGFR wild-type lung adenocarcinoma. Commun Biol 2024; 7:1416. [PMID: 39478024 PMCID: PMC11525656 DOI: 10.1038/s42003-024-07117-1] [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: 02/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Epidermal growth factor receptor wild type lung adenocarcinoma (EGFRWT LUAD) still has limited treatment options and unsatisfactory clinical outcomes. Ferroptosis, as a form of cell death, has been reported to play a dual role in regulating tumor cell survival. In this study, we constructed a 3-ferroptosis-gene signature, FeSig, and verified its accuracy and efficacy in predicting EGFRWT LUAD prognosis at both the RNA and protein levels. Patients with higher FeSig scores were found to have worse clinical outcomes. Additionally, we explored the relationship between FeSig and tumor microenvironment, revealing that enhanced interactions between fibroblasts and tumor cells in FeSighigh patients causing tumor resistance to ferroptosis. To address this challenge, we screened potential drugs from NCI-60 (The US National Cancer Institute 60 human tumour cell line anticancer drug screen) and Connectivity map database, ultimately identifying 6-mercatopurine (6-MP) as a promising candidate. Both in vitro and in vivo experiments demonstrated its efficacy in treating FeSighigh EGFRWT LUAD tumor models. In summary, we develop a novel FeSig for predicting prognosis and guiding drug application.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hongwei Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Changqing Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Yang
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qingsong Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| |
Collapse
|
43
|
Meng F, Li H, Jin R, Yang A, Luo H, Li X, Wang P, Zhao Y, Chervova O, Tang K, Cheng S, Hu B, Li Y, Sheng J, Yang F, Carbone D, Chen K, Wang J. Spatial immunogenomic patterns associated with lymph node metastasis in lung adenocarcinoma. Exp Hematol Oncol 2024; 13:106. [PMID: 39468696 PMCID: PMC11514955 DOI: 10.1186/s40164-024-00574-8] [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: 07/30/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) with lymph node (LN) metastasis is linked to poor prognosis, yet the underlying mechanisms remain largely undefined. This study aimed to elucidate the immunogenomic landscape associated with LN metastasis in LUAD. METHODS We employed broad-panel next-generation sequencing (NGS) on a cohort of 257 surgically treated LUAD patients to delineate the molecular landscape of primary tumors and identify actionable driver-gene alterations. Additionally, we used multiplex immunohistochemistry (mIHC) on a propensity score-matched cohort, which enabled us to profile the immune microenvironment of primary tumors in detail while preserving cellular metaclusters, interactions, and neighborhood functional units. By integrating data from NGS and mIHC, we successfully identified spatial immunogenomic patterns and developed a predictive model for LN metastasis, which was subsequently validated independently. RESULTS Our analysis revealed distinct immunogenomic alteration patterns associated with LN metastasis stages. Specifically, we observed increased mutation frequencies in genes such as PIK3CG and ATM in LN metastatic primary tumors. Moreover, LN positive primary tumors exhibited a higher presence of macrophage and regulatory T cell metaclusters, along with their enriched neighborhood units (p < 0.05), compared to LN negative tumors. Furthermore, we developed a novel predictive model for LN metastasis likelihood, designed to inform non-surgical treatment strategies, optimize personalized therapy plans, and potentially improve outcomes for patients who are ineligible for surgery. CONCLUSIONS This study offers a comprehensive analysis of the genetic and immune profiles in LUAD primary tumors with LN metastasis, identifying key immunogenomic patterns linked to metastatic progression. The predictive model derived from these insights marks a substantial advancement in personalized treatment, underscoring its potential to improve patient management.
Collapse
Affiliation(s)
- Fanjie Meng
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Ruoyi Jin
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Airong Yang
- Kanghui Biotechnology Co., Ltd, Shenyang, China
| | - Hao Luo
- Cancer Center, Daping Hospital Army Medical University, Chongqing, China
| | - Xiao Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yaxing Zhao
- Infinity Scope Biotechnology Co., Ltd., Hangzhou, China
| | - Olga Chervova
- University College London Cancer Institute, University College London, London, UK
| | - Kaicheng Tang
- Infinity Scope Biotechnology Co., Ltd., Hangzhou, China
| | - Sida Cheng
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - Jianpeng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China
- Chinese Institutes for Medical Research, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China
| | - David Carbone
- James Thoracic Oncology Center, Ohio State University, Columbus, USA
| | - Kezhong Chen
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China.
- Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-Small Cell Lung Cancer, Peking University People's Hospital, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
| | - Jun Wang
- Department of Thoracic Surgery, Institution of Thoracic Oncology, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, Xicheng District, China.
| |
Collapse
|
44
|
Zhang H, Liu C, Wang S, Wang Q, Feng X, Jiang H, Xiao L, Luo C, Zhang L, Hou F, Zhou M, Deng Z, Li H, Zhang Y, Su X, Li G. Proteogenomic analysis of air-pollution-associated lung cancer reveals prevention and therapeutic opportunities. eLife 2024; 13:RP95453. [PMID: 39432560 PMCID: PMC11493407 DOI: 10.7554/elife.95453] [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] [Indexed: 10/23/2024] Open
Abstract
Air pollution significantly impacts lung cancer progression, but there is a lack of a comprehensive molecular characterization of clinical samples associated with air pollution. Here, we performed a proteogenomic analysis of lung adenocarcinoma (LUAD) in 169 female never-smokers from the Xuanwei area (XWLC cohort), where coal smoke is the primary contributor to the high lung cancer incidence. Genomic mutation analysis revealed XWLC as a distinct subtype of LUAD separate from cases associated with smoking or endogenous factors. Mutational signature analysis suggested that Benzo[a]pyrene (BaP) is the major risk factor in XWLC. The BaP-induced mutation hotspot, EGFR-G719X, was present in 20% of XWLC which endowed XWLC with elevated MAPK pathway activations and worse outcomes compared to common EGFR mutations. Multi-omics clustering of XWLC identified four clinically relevant subtypes. These subgroups exhibited distinct features in biological processes, genetic alterations, metabolism demands, immune landscape, and radiomic features. Finally, MAD1 and TPRN were identified as novel potential therapeutic targets in XWLC. Our study provides a valuable resource for researchers and clinicians to explore prevention and treatment strategies for air-pollution-associated lung cancers.
Collapse
Affiliation(s)
- Honglei Zhang
- Center for Scientific Research, Yunnan University of Chinese MedicineKunmingChina
| | - Chao Liu
- Department of Nuclear Medicine, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| | - Shuting Wang
- Department of Thoracic Surgery II, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| | - Qing Wang
- Department of Oncology, Qujing First People’s HospitalKumingChina
| | - Xu Feng
- Center for Scientific Research, Yunnan University of Chinese MedicineKunmingChina
| | - Huawei Jiang
- Department of Ophthalmology, Second People's Hospital of Yunnan ProvinceKunmingChina
| | - Li Xiao
- Department of Oncology, Qujing First People’s HospitalKumingChina
| | - Chao Luo
- Department of Thoracic Surgery II, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| | - Lu Zhang
- Department of Nuclear Medicine, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| | - Fei Hou
- Department of Nuclear Medicine, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| | - Minjun Zhou
- Department of Family Medicine, Community Health Service CenterKunmingChina
| | - Zhiyong Deng
- Department of Nuclear Medicine, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| | - Heng Li
- Department of Thoracic Surgery II, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| | - Yong Zhang
- Department of Nephrology, Institutes for Systems Genetics, Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan UniversityChengduChina
| | - Xiaosan Su
- Center for Scientific Research, Yunnan University of Chinese MedicineKunmingChina
| | - Gaofeng Li
- Department of Thoracic Surgery II, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingChina
| |
Collapse
|
45
|
Li H, Huang Z, Guo C, Wang Y, Li B, Wang S, Bai N, Chen H, Xue J, Wang D, Zheng Z, Bing Z, Song Y, Xu Y, Huang G, Yu X, Li R, Fung KL, Li J, Song L, Zhu Z, Liu S, Liang N, Li S. Super multiple primary lung cancers harbor high-frequency BRAF and low-frequency EGFR mutations in the MAPK pathway. NPJ Precis Oncol 2024; 8:229. [PMID: 39384982 PMCID: PMC11464572 DOI: 10.1038/s41698-024-00726-3] [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: 03/26/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
The incidence of multiple primary lung cancer (MPLC) is increasing, with some of our surgical patients exhibiting numerous lesions. We defined lung cancer with five or more primary lesions as super MPLCs. Elucidating the genomic characteristics of this special MPLC subtype can help reduce disease burden and understand tumor evolution. In our cohort of synchronous super early-stage MPLCs (PUMCH-ssesMPLC), whole-exome sequencing on 130 resected malignant specimens from 18 patients provided comprehensive super-MPLC genomic landscapes. Mutations are enriched in PI3k-Akt and MAPK pathways. Their BRAF mutation frequency (31.5%) is significantly higher than MPLC with fewer lesions and early-stage single-lesion cancer, while EGFR mutations are significantly fewer (13.8%). As lesion counts increase, BRAF mutations gradually become dominant. Also, invasive lesions more tend to have classic super-MPLC mutation patterns. High-frequency BRAF mutations, especially Class II, and low-frequency EGFR mutations could be a reason for the limited effectiveness of targeted therapy in super-MPLC patients.
Collapse
Affiliation(s)
- Haochen Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Zhicheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Bowen Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Sha Wang
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, 210032, China
| | - Na Bai
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, 210032, China
| | - Hanlin Chen
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, 210032, China
| | - Jianchao Xue
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Daoyun Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhibo Zheng
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of International Medical Services, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yang Song
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuan Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guanghua Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiaoqing Yu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruirui Li
- Department of Thoracic Surgery, Aviation General Hospital, Beijing, 100025, China
| | | | - Ji Li
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lan Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziwei Zhu
- Zhenyuan (Tianjin) Medical Technology Co. Ltd., Tianjin, 300385, China
| | - Songtao Liu
- Zhenyuan (Tianjin) Medical Technology Co. Ltd., Tianjin, 300385, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
46
|
Wei J, Wang X, Guo H, Zhang L, Shi Y, Wang X. Subclassification of lung adenocarcinoma through comprehensive multi-omics data to benefit survival outcomes. Comput Biol Chem 2024; 112:108150. [PMID: 39018587 DOI: 10.1016/j.compbiolchem.2024.108150] [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: 12/25/2023] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
OBJECTIVES Lung adenocarcinoma (LUAD) is the most common subtype of non-small cell lung cancer. Understanding the molecular mechanisms underlying tumor progression is of great clinical significance. This study aims to identify novel molecular markers associated with LUAD subtypes, with the goal of improving the precision of LUAD subtype classification. Additionally, optimization efforts are directed towards enhancing insights from the perspective of patient survival analysis. MATERIALS AND METHODS We propose an innovative feature-selection approach that focuses on LUAD classification, which is comprehensive and robust. The proposed method integrates multi-omics data from The Cancer Genome Atlas (TCGA) and leverages a synergistic combination of max-relevance and min-redundancy, least absolute shrinkage and selection operator, and Boruta algorithms. These selected features were deployed in six machine-learning classifiers: logistic regression, random forest, support vector machine, naive Bayes, k-Nearest Neighbor, and XGBoost. RESULTS The proposed approach achieved an area under the receiver operating characteristic curve (AUC) of 0.9958 for LR. Notably, the accuracy and AUC of a composite model incorporating copy number, methylation, as well as RNA- sequencing data for expression of exons, genes, and miRNA mature strands surpassed the accuracy and AUC metrics of models with single-omics data or other multi-omics combinations. Survival analyses, revealed the SVM classifier to elicit optimal classification, outperforming that achieved by TCGA. To enhance model interpretability, SHapley Additive exPlanations (SHAP) values were utilized to elucidate the impact of each feature on the predictions. Gene Ontology (GO) enrichment analysis identified significant biological processes, molecular functions, and cellular components associated with LUAD subtypes. CONCLUSION In summary, our feature selection process, based on TCGA multi-omics data and combined with multiple machine learning classifiers, proficiently identifies molecular subtypes of lung adenocarcinoma and their corresponding significant genes. Our method could enhance the early detection and diagnosis of LUAD, expedite the development of targeted therapies and, ultimately, lengthen patient survival.
Collapse
Affiliation(s)
| | - Xin Wang
- Qingdao University, Qingdao, China
| | | | - Ling Zhang
- Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Yao Shi
- Qingdao University, Qingdao, China.
| | | |
Collapse
|
47
|
Mao S, Wang Y, Chao N, Zeng L, Zhang L. Integrated analysis of single-cell RNA-seq and bulk RNA-seq reveals immune suppression subtypes and establishes a novel signature for determining the prognosis in lung adenocarcinoma. Cell Oncol (Dordr) 2024; 47:1697-1713. [PMID: 38616208 DOI: 10.1007/s13402-024-00948-4] [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] [Accepted: 04/07/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the most common histological type of lung cancer with lower survival rates. Recent advancements in targeted therapies and immunotherapies targeting immune checkpoints have achieved remarkable success, there is still a large percentage of LUAD that lacks available therapeutic options. Due to tumor heterogeneity, the diagnosis and treatment of LUAD are challenging. Exploring the biology of LUAD and identifying new biomarker and therapeutic targets options are essential. METHOD We performed single-cell RNA sequencing (scRNA-seq) of 6 paired primary and adjacent LUAD tissues, and integrative omics analysis of the scRNA-seq, bulk RNA-seq and whole-exome sequencing data revealed molecular subtype characteristics. Our experimental results confirm that CDC25C gene can serve as a potential marker for poor prognosis in LUAD. RESULTS We investigated aberrant gene expression in diverse cell types in LUAD via the scRNA-seq data. Moreover, multi-omics clustering revealed four subgroups defined by transcriptional profile and molecular subtype 4 (MS4) with poor survival probability, and immune cell infiltration signatures revealed that MS4 tended to be the immunosuppressive subtype. Our study revealed that the CDC25C gene can be a distinct prognostic biomarker that indicates immune infiltration levels and response to immunotherapy in LUAD patients. Our experimental results concluded that CDC25C expression affects lung cancer cell invasion and migration, might play a key role in regulating Epithelial-Mesenchymal Transition (EMT) pathways. CONCLUSIONS Our multi-omics result revealed a comprehensive set of molecular attributes associated with prognosis-related genes in LUAD at the cellular and tissue level. Identification of a subtype of immunosuppressive TME and prognostic signature for LUAD. We identified the cell cycle regulation gene CDC25C affects lung cancer cell invasion and migration, which can be used as a potential biomarker for LUAD.
Collapse
Affiliation(s)
- Shengqiang Mao
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yilong Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ningning Chao
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lingyan Zeng
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
48
|
Caballé-Perez E, Cabrera-Miranda L, Arrieta O. Exploring EGFR Mutations in Resected Stage I-III NSCLC in Low- and Middle-Income Countries: Bridging the Gap in Global Cancer Care. J Thorac Oncol 2024; 19:1367-1369. [PMID: 39370217 DOI: 10.1016/j.jtho.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 10/08/2024]
Affiliation(s)
- Enrique Caballé-Perez
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Ciudad de México, México; Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Ciudad de México, México.
| | - Luis Cabrera-Miranda
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Ciudad de México, México
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Ciudad de México, México; Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Ciudad de México, México
| |
Collapse
|
49
|
Zeng L, Zhang L, Li L, Liao X, Yin C, Zhang L, Chen X, Sun J. RNA sequencing identifies lung cancer lineage and facilitates drug repositioning. PeerJ 2024; 12:e18159. [PMID: 39346064 PMCID: PMC11430167 DOI: 10.7717/peerj.18159] [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: 05/27/2024] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Recent breakthrough therapies have improved survival rates in non-small cell lung cancer (NSCLC), but a paradigm for prospective confirmation is still lacking. Patientdatasets were mainly downloaded from TCGA, CPTAC and GEO. We conducted downstream analysis by collecting metagenes and generated 42-gene subtype classifiers to elucidate biological pathways. Subsequently, scRNA, eRNA, methylation, mutation, and copy number variation were depicted from a phenotype perspective. Enhancing the clinical translatability of molecular subtypes, preclinical models including CMAP, CCLE, and GDSC were utilized for drug repositioning. Importantly, we verified the presence of previously described three phenotypes including bronchioid, neuroendocrine, and squamoid. Poor prognosis was seen in squamoid and neuroendocrine clusters for treatment-naive and immunotherapy populations. The neuroendocrine cluster was dominated by STK11 mutations and 14q13.3 amplifications, whose related methylated loci are predictive of immunotherapy. And the greatest therapeutic potential lies in the bronchioid cluster. We further estimated the relative cell abundance of the tumor microenvironment (TME), specific cell types could be reflected among three clusters. Meanwhile, the higher portion of immune cell infiltration belonged to bronchioid and squamoid, not the neuroendocrine cluster. In drug repositioning, MEK inhibitors resisted bronchioid but were squamoid-sensitive. To conceptually validate compounds/targets, we employed RNA-seq and CCK-8/western blot assays. Our results indicated that dinaciclib and alvocidib exhibited similar activity and sensitivity in the neuroendocrine cluster. Also, a lineage factor named KLF5 recognized by inferred transcriptional factors activity could be suppressed by verteporfin.
Collapse
Affiliation(s)
- Longjin Zeng
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Longyao Zhang
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| | - Lingchen Li
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| | - Xingyun Liao
- Affiliated Tumor Hospital, Department of Oncology, Chongqing, China
| | - Chenrui Yin
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| | - Lincheng Zhang
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiewan Chen
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Jianguo Sun
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| |
Collapse
|
50
|
Lai GGY, Tan DSW. Lung cancer screening in never smokers. Curr Opin Oncol 2024:00001622-990000000-00212. [PMID: 39258345 DOI: 10.1097/cco.0000000000001099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
PURPOSE OF REVIEW Low-dose computed tomography (LDCT) lung cancer screening has been established in smokers, but its role in never smokers remains unclear. The differences in lung cancer biology between smokers and nonsmokers highlight the importance of a discriminated approach. This overview focuses on the emerging data and implementation challenges for LDCT screening in nonsmokers. RECENT FINDINGS The first LDCT screening study in nonsmokers enriched with risk factors demonstrated a lung cancer detection rate double that of the phase 3 trials in smokers. The relative risk of lung cancer detected by LDCT has also been found to be similar amongst female never smokers and male ever smokers in Asia. Majority of lung cancers detected through LDCT screening are stage 0/1, leading to concerns of overdiagnosis. Risk prediction models to enhance individual selection and nodule management could be useful to enhance the utility of LDCT screening in never smokers. SUMMARY With appropriate risk stratification, LDCT screening in never smokers may attain similar efficacy as compared to smokers. A global effort is needed to generate evidence surrounding optimal screening strategies, as well as health and economic benefits to determine the suitability of widespread implementation.
Collapse
Affiliation(s)
- Gillianne G Y Lai
- Division of Medical Oncology, National Cancer Centre Singapore
- Duke-NUS Medical School
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore
- Duke-NUS Medical School
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| |
Collapse
|