Ma Ke, Xu Jie, Wang Congyue, Cao Xu, Yu Wenjie, Xi Jingjing, Zhang Xuan, Zhan Jiamin, Liu Yang, Yu Aoyang, Liu Shuhan, Liu Yanhua, Chen Chong, Mai Xiaoli
Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, Jiangsu, China.
Institute of Hematology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
Front Oncol. 2025 Jul 21;15:1590216. doi: 10.3389/fonc.2025.1590216. eCollection 2025.
The development of high-throughput sequencing technologies and targeted therapeutic strategies has significantly improved the prognosis of lung adenocarcinoma (LUAD) patients with sensitive gene mutations. However, patients harboring rare or no actionable mutations were rarely benefit from these targeted therapies. This study aimed to identify novel molecular subtypes and construct a prognostic signature to enhance the stratification of LUAD prognosis.
Novel molecular subtypes of LUAD patients were identified by applying 10 distinct clustering algorithms on multi-omics data. Single-cell RNA-sequencing (scRNA-seq) data were integrated to characterize subtype-specific immune microenvironments. A multi-omics and machine learning-driven prognostic signature (MO-MLPS) was constructed in The Cancer Genome Atlas (TCGA) LUAD dataset using ten machine learning algorithms and subsequently validated across six independent datasets from the Gene Expression Omnibus (GEO) database. The robustness of the model was assessed using the concordance index (C-index), Kaplan-Meier survival analyses, receiver operating characteristic (ROC) curves, and both univariate and multivariate Cox regression analyses. We further confirmed the effects of ANLN knockdown and the expression of a domain-negative anillin protein (dnANLN) via western blotting, cell proliferation assays, flow cytometry, and transwell migration assays .
Our analysis revealed that the novel molecular subtypes exhibited differences in prognoses, biological functions, and immune infiltration profiles in LUAD. The MO-MLPS was successfully established and validated across TCGA-LUAD cohorts, six independent GEO datasets, and their composite meta-cohort. Higher risk scores from the MO-MLPS correlated with poorer prognosis in LUAD, with AUC values exceeding 0.5 at 1, 3, and 5 years across various cohorts. The signature outperformed 49 previously published prognostic signatures. Furthermore, patients classified as high risk exhibited significantly worse overall and progression-free survival than those classified as low risk. Notably, ANLN knockdown and dnANLN expression significantly inhibited cell proliferation and migration and enhanced the efficacy of docetaxel.
A comprehensive analysis of multi-omics data redefines the molecular subtype of LUAD patients. The MO-MLPS derived from subtype characteristics has the potential to serve as a clinically valuable prognostic tool. Furthermore, ANLN emerges as a promising novel therapeutic target in the treatment of LUAD.
高通量测序技术和靶向治疗策略的发展显著改善了具有敏感基因突变的肺腺癌(LUAD)患者的预后。然而,携带罕见或无可用靶点突变的患者很少能从这些靶向治疗中获益。本研究旨在识别新的分子亚型并构建预后特征以加强LUAD预后的分层。
通过对多组学数据应用10种不同的聚类算法来识别LUAD患者的新分子亚型。整合单细胞RNA测序(scRNA-seq)数据以表征亚型特异性免疫微环境。使用十种机器学习算法在癌症基因组图谱(TCGA)LUAD数据集中构建多组学和机器学习驱动的预后特征(MO-MLPS),随后在来自基因表达综合数据库(GEO)的六个独立数据集中进行验证。使用一致性指数(C-index)、Kaplan-Meier生存分析、受试者工作特征(ROC)曲线以及单变量和多变量Cox回归分析来评估模型的稳健性。我们通过蛋白质免疫印迹、细胞增殖试验、流式细胞术和Transwell迁移试验进一步证实了ANLN敲低和显性负性膜收缩蛋白(dnANLN)的表达效果。
我们的分析表明,新的分子亚型在LUAD的预后、生物学功能和免疫浸润特征方面存在差异。MO-MLPS在TCGA-LUAD队列、六个独立的GEO数据集及其复合meta队列中成功建立并得到验证。MO-MLPS的较高风险评分与LUAD的较差预后相关,在各个队列中1年、3年和5年的AUC值均超过0.5。该特征优于先前发表的49种预后特征。此外,被分类为高风险的患者的总生存期和无进展生存期明显比被分类为低风险的患者差。值得注意的是,ANLN敲低和dnANLN表达显著抑制细胞增殖和迁移,并增强了多西他赛的疗效。
对多组学数据的综合分析重新定义了LUAD患者的分子亚型。源自亚型特征的MO-MLPS有潜力作为一种具有临床价值的预后工具。此外,ANLN成为LUAD治疗中有前景的新治疗靶点。