Suppr超能文献

剖析EGFR/TP53共突变非小细胞肺癌中的免疫逃逸和治疗耐药机制:对靶向治疗和免疫治疗策略的启示

Dissecting the immune evasion and therapeutic resistance mechanisms in EGFR/TP53 co-mutated non-small cell lung cancer: implications for targeted and immunotherapy strategies.

作者信息

Shi Haiyan, Xu Kun, Kong Xueying, Xie Weining, Chen Yingying, He Ding, Cheng Zufu, Huo Xianshan, Gao Ke, Song Mingshuang, Tian Ning

机构信息

Department of Pathology, Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, Guangdong, China.

Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.

出版信息

Front Immunol. 2025 Aug 29;16:1652213. doi: 10.3389/fimmu.2025.1652213. eCollection 2025.

Abstract

BACKGROUND

Although precision-targeted therapies and tyrosine kinase inhibitors (TKIs) have significantly improved outcomes in non-small-cell lung cancer (NSCLC), patients with EGFR-mutant NSCLC with concurrent TP53 mutations often develop drug resistance and experience poor clinical outcomes. This study aims to investigate the molecular mechanisms underlying this aggressive subtype using single-cell RNA sequencing.

METHODS

Formalin-fixed paraffin-embedded (FFPE) tumor samples were obtained from 40 hospitalized NSCLC patients. Somatic mutation profiles were determined using a targeted 23-gene next-generation sequencing (NGS) panel. Four samples harboring concurrent EGFR and TP53 mutations were selected for single-cell transcriptomic profiling using the 10x Genomics platform.

RESULTS

Two dominant malignant epithelial cell populations were identified: C1_EGFR, associated with proliferation and invasion, and C2_STAT1, linked to immunosuppression and drug resistance. These tumor subtypes cooperatively drive CD8 T cell exhaustion through the MDK-(ITGA4+ITGB1), MIF-(CD74+CXCR4), and TGF-β signaling pathways. In addition, antigen-presenting cancer-associated fibroblasts (apCAFs) recruit regulatory T cells via the CCL5-CCR4 axis, collectively establishing an immune-excluded tumor microenvironment. Mechanistically, a STAT1/ETS1-centered transcriptional program regulates the expression of key immunosuppressive (e.g., MDK, MIF, TGFB1) and resistance-associated genes (e.g., ERBB2, JAK2).

CONCLUSION

These findings reveal a coordinated transcriptional network that promotes immune evasion and therapeutic resistance in EGFR/TP53 co-mutated NSCLC. Targeting the STAT1/ETS1 axis, in combination with EGFR-TKIs or immune checkpoint inhibitors, may provide a novel strategy to overcome resistance and improve patient outcomes. Further validation in larger patient cohorts and functional studies is warranted to confirm these observations and support clinical translation.

摘要

背景

尽管精准靶向治疗和酪氨酸激酶抑制剂(TKIs)显著改善了非小细胞肺癌(NSCLC)的治疗效果,但伴有TP53突变的EGFR突变NSCLC患者常出现耐药且临床预后较差。本研究旨在通过单细胞RNA测序探究这种侵袭性亚型背后的分子机制。

方法

从40例住院NSCLC患者中获取福尔马林固定石蜡包埋(FFPE)肿瘤样本。使用靶向23基因的二代测序(NGS) panel确定体细胞突变谱。选择4例同时存在EGFR和TP53突变的样本,使用10x Genomics平台进行单细胞转录组分析。

结果

鉴定出两个主要的恶性上皮细胞群体:与增殖和侵袭相关的C1_EGFR,以及与免疫抑制和耐药相关的C2_STAT1。这些肿瘤亚型通过MDK-(ITGA4+ITGB1)、MIF-(CD74+CXCR4)和TGF-β信号通路协同驱动CD8 T细胞耗竭。此外,抗原呈递癌相关成纤维细胞(apCAFs)通过CCL5-CCR4轴募集调节性T细胞,共同建立免疫排斥的肿瘤微环境。机制上,以STAT1/ETS1为中心转录程序调节关键免疫抑制(如MDK、MIF、TGFB1)和耐药相关基因(如ERBB2、JAK2)的表达。

结论

这些发现揭示了一个协调的转录网络,该网络促进EGFR/TP53共突变NSCLC中的免疫逃逸和治疗耐药。靶向STAT1/ETS1轴,联合EGFR-TKIs或免疫检查点抑制剂,可能提供一种克服耐药并改善患者预后的新策略。需要在更大的患者队列中进一步验证并进行功能研究,以证实这些观察结果并支持临床转化。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/02e0/12425899/e069693eb3e9/fimmu-16-1652213-g001.jpg

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍。

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

文档翻译

学术文献翻译模型,支持多种主流文档格式。

立即体验