Multi-Omics and Single-Cell Insights Reveal a Lysophosphatidic Acid (LPA)-Mediated Resistant Mechanism to Third Generation EGFR-TKI in Non-Small Cell Lung Cancer
Ruyun Gao, Ning Lou, Sheng Yang, Mengwei Yang, Guangyu Fan, Liyuan Dai, Le Tang, Jiarui Yao, Xiaohong Han, Yuankai Shi
{"title":"Multi-Omics and Single-Cell Insights Reveal a Lysophosphatidic Acid (LPA)-Mediated Resistant Mechanism to Third Generation EGFR-TKI in Non-Small Cell Lung Cancer","authors":"Ruyun Gao, Ning Lou, Sheng Yang, Mengwei Yang, Guangyu Fan, Liyuan Dai, Le Tang, Jiarui Yao, Xiaohong Han, Yuankai Shi","doi":"10.1158/1078-0432.ccr-25-0993","DOIUrl":null,"url":null,"abstract":"Introduction: Third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of EGFR-sensitive-mutant non-small cell lung cancer (NSCLC). However, acquired resistance remains a significant challenge. This study investigates the metabolic mechanisms driving third-generation EGFR-TKI resistance. Methods: We conducted plasma metabolomics analysis on 216 longitudinal samples from 186 NSCLC patients enrolled in the clinical trial of rezivertinib (NCT03386955). Additionally, multi-omics profiling of rezivertinib-resistant cell lines, functional in vitro experiments, and single-cell RNA sequencing (scRNA-seq) analyses of 215 NSCLC patients were integrated to reveal underlying mechanisms. Results: Non-responder patients exhibited elevated glycerophospholipids and dysregulated lysophospholipid (LPL) metabolism. Unsupervised clustering identified two patient subgroups, with Cluster 1 (characterized by high LPL levels) associated with poorer survival (P = 0.022). Metabolite-based predictive model achieved robust performance (AUC: 0.7762 [training], 0.7485 [test]). Longitudinal analyses demonstrated LPLs and lysophosphatidic acid (LPA) accumulation during the resistance process. Integrated multi-omics analyses highlighted epithelial-mesenchymal transition (EMT) and glycerophospholipid reprogramming in rezivertinib-resistant cells. Functional assays confirmed that LPA promoted cell migration and invasion, and attenuated the efficacy of third-generation EGFR-TKI, while disruption of the LPA-LPAR signaling axis reversed LPA-mediated resistance. scRNA-seq identified an LPA-secreting malignant subset (cluster c4), characterized by enhanced EMT activation and extensive microenvironmental crosstalk through Wnt, TGF-β, and ECM signals. Conclusions: Our study highlights the pivotal role of LPA-mediated signaling and metabolic reprogramming in third-generation EGFR-TKI resistance. Targeting LPA production or its downstream pathways may offer novel therapeutic strategies to overcome resistance. This study provides critical metabolic insights for managing EGFR-sensitive-mutant NSCLC.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"132 1","pages":""},"PeriodicalIF":10.2000,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Cancer Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1158/1078-0432.ccr-25-0993","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0
Abstract
Introduction: Third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of EGFR-sensitive-mutant non-small cell lung cancer (NSCLC). However, acquired resistance remains a significant challenge. This study investigates the metabolic mechanisms driving third-generation EGFR-TKI resistance. Methods: We conducted plasma metabolomics analysis on 216 longitudinal samples from 186 NSCLC patients enrolled in the clinical trial of rezivertinib (NCT03386955). Additionally, multi-omics profiling of rezivertinib-resistant cell lines, functional in vitro experiments, and single-cell RNA sequencing (scRNA-seq) analyses of 215 NSCLC patients were integrated to reveal underlying mechanisms. Results: Non-responder patients exhibited elevated glycerophospholipids and dysregulated lysophospholipid (LPL) metabolism. Unsupervised clustering identified two patient subgroups, with Cluster 1 (characterized by high LPL levels) associated with poorer survival (P = 0.022). Metabolite-based predictive model achieved robust performance (AUC: 0.7762 [training], 0.7485 [test]). Longitudinal analyses demonstrated LPLs and lysophosphatidic acid (LPA) accumulation during the resistance process. Integrated multi-omics analyses highlighted epithelial-mesenchymal transition (EMT) and glycerophospholipid reprogramming in rezivertinib-resistant cells. Functional assays confirmed that LPA promoted cell migration and invasion, and attenuated the efficacy of third-generation EGFR-TKI, while disruption of the LPA-LPAR signaling axis reversed LPA-mediated resistance. scRNA-seq identified an LPA-secreting malignant subset (cluster c4), characterized by enhanced EMT activation and extensive microenvironmental crosstalk through Wnt, TGF-β, and ECM signals. Conclusions: Our study highlights the pivotal role of LPA-mediated signaling and metabolic reprogramming in third-generation EGFR-TKI resistance. Targeting LPA production or its downstream pathways may offer novel therapeutic strategies to overcome resistance. This study provides critical metabolic insights for managing EGFR-sensitive-mutant NSCLC.
期刊介绍:
Clinical Cancer Research is a journal focusing on groundbreaking research in cancer, specifically in the areas where the laboratory and the clinic intersect. Our primary interest lies in clinical trials that investigate novel treatments, accompanied by research on pharmacology, molecular alterations, and biomarkers that can predict response or resistance to these treatments. Furthermore, we prioritize laboratory and animal studies that explore new drugs and targeted agents with the potential to advance to clinical trials. We also encourage research on targetable mechanisms of cancer development, progression, and metastasis.