{"title":"ROS1突变通过MYC促进免疫抑制肿瘤微环境,从而导致头颈癌的免疫逃避。","authors":"Chao Fang, Qin Zhang, Rui Fang, Ying Li, Jing Bai, Xiaojing Huang, Jingting Lu, Dongsheng Chen, Yanxiang Zhang, Zuhong Chen","doi":"10.20517/cdr.2025.124","DOIUrl":null,"url":null,"abstract":"<p><p><b>Aim:</b> Immune checkpoint inhibitors (ICIs) have transformed cancer therapy; however, their efficacy in head and neck cancer (HNC) remains limited, with only a minority of patients achieving durable responses. Understanding the molecular mechanisms underlying ICI resistance in HNC is therefore crucial. <b>Methods:</b> We conducted an integrative analysis of genomic, transcriptomic, and clinical data from 139 ICI-treated HNC patients (MSKCC cohort) and 502 treatment-naïve HNC cases (TCGA cohort). <i>ROS1</i> mutation status, tumor mutational burden (TMB), neoantigen load, immune cell infiltration (via CIBERSORT), and immune-related gene expression were evaluated. Gene set enrichment analysis (GSEA) was performed to identify dysregulated pathways. Survival outcomes were assessed using Kaplan-Meier analysis and Cox regression, with statistical significance defined as <i>P</i> < 0.05. <b>Results:</b> Patients harboring <i>ROS1</i> mutations exhibited significantly poorer outcomes following ICI therapy, with shorter median overall survival [OS: 5.0 <i>vs.</i> 11.0 months, hazard ratio (HR) = 3.22, 95%CI: 1.26-8.19, <i>P</i> = 0.011] compared to <i>ROS1</i> wild-type counterparts. Multivariate analysis confirmed <i>ROS1</i> mutation as an independent predictor of poor OS in ICI-treated patients (HR = 4.78, 95%CI: 1.70-13.43, <i>P</i> = 0.003). In contrast, <i>ROS1</i> mutations showed no prognostic significance in the treatment-naïve TCGA-HNC cohort (<i>P</i> = 0.26), confirming their role as a predictive (not prognostic) biomarker for ICI response. Interestingly, despite exhibiting higher TMB and neoantigen levels, <i>ROS1</i>-mutant patients showed inferior survival, underscoring the context-dependent limitations of TMB as a predictive biomarker. Mechanistically, <i>ROS1</i>-mutant tumors displayed an immunosuppressive tumor microenvironment characterized by diminished CD8<sup>+</sup> T cell infiltration, attenuated interferon-γ signaling, and downregulation of immune-related genes (<i>CXCL9</i>, <i>CXCL10</i>, <i>IFNG</i>, <i>PD-L1</i>). GSEA revealed enrichment of <i>MYC</i> pathway activity in <i>ROS1</i>-mutant tumors, which suppressed antigen presentation and T cell activation pathways. <b>Conclusion:</b> <i>ROS1</i> mutations drive ICI resistance in HNC by promoting an immunosuppressive TME via <i>MYC</i>-mediated transcriptional reprogramming, impairing antigen presentation and T cell function. Incorporating <i>ROS1</i> status into biomarker panels may improve patient stratification and guide combinatorial therapies targeting both immune evasion and oncogenic pathways.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"42"},"PeriodicalIF":4.6000,"publicationDate":"2025-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12378350/pdf/","citationCount":"0","resultStr":"{\"title\":\"<i>ROS1</i> mutations promote an immunosuppressive tumor microenvironment via <i>MYC</i> to confer immune evasion in head and neck cancer.\",\"authors\":\"Chao Fang, Qin Zhang, Rui Fang, Ying Li, Jing Bai, Xiaojing Huang, Jingting Lu, Dongsheng Chen, Yanxiang Zhang, Zuhong Chen\",\"doi\":\"10.20517/cdr.2025.124\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p><b>Aim:</b> Immune checkpoint inhibitors (ICIs) have transformed cancer therapy; however, their efficacy in head and neck cancer (HNC) remains limited, with only a minority of patients achieving durable responses. Understanding the molecular mechanisms underlying ICI resistance in HNC is therefore crucial. <b>Methods:</b> We conducted an integrative analysis of genomic, transcriptomic, and clinical data from 139 ICI-treated HNC patients (MSKCC cohort) and 502 treatment-naïve HNC cases (TCGA cohort). <i>ROS1</i> mutation status, tumor mutational burden (TMB), neoantigen load, immune cell infiltration (via CIBERSORT), and immune-related gene expression were evaluated. Gene set enrichment analysis (GSEA) was performed to identify dysregulated pathways. Survival outcomes were assessed using Kaplan-Meier analysis and Cox regression, with statistical significance defined as <i>P</i> < 0.05. <b>Results:</b> Patients harboring <i>ROS1</i> mutations exhibited significantly poorer outcomes following ICI therapy, with shorter median overall survival [OS: 5.0 <i>vs.</i> 11.0 months, hazard ratio (HR) = 3.22, 95%CI: 1.26-8.19, <i>P</i> = 0.011] compared to <i>ROS1</i> wild-type counterparts. Multivariate analysis confirmed <i>ROS1</i> mutation as an independent predictor of poor OS in ICI-treated patients (HR = 4.78, 95%CI: 1.70-13.43, <i>P</i> = 0.003). In contrast, <i>ROS1</i> mutations showed no prognostic significance in the treatment-naïve TCGA-HNC cohort (<i>P</i> = 0.26), confirming their role as a predictive (not prognostic) biomarker for ICI response. Interestingly, despite exhibiting higher TMB and neoantigen levels, <i>ROS1</i>-mutant patients showed inferior survival, underscoring the context-dependent limitations of TMB as a predictive biomarker. Mechanistically, <i>ROS1</i>-mutant tumors displayed an immunosuppressive tumor microenvironment characterized by diminished CD8<sup>+</sup> T cell infiltration, attenuated interferon-γ signaling, and downregulation of immune-related genes (<i>CXCL9</i>, <i>CXCL10</i>, <i>IFNG</i>, <i>PD-L1</i>). GSEA revealed enrichment of <i>MYC</i> pathway activity in <i>ROS1</i>-mutant tumors, which suppressed antigen presentation and T cell activation pathways. <b>Conclusion:</b> <i>ROS1</i> mutations drive ICI resistance in HNC by promoting an immunosuppressive TME via <i>MYC</i>-mediated transcriptional reprogramming, impairing antigen presentation and T cell function. Incorporating <i>ROS1</i> status into biomarker panels may improve patient stratification and guide combinatorial therapies targeting both immune evasion and oncogenic pathways.</p>\",\"PeriodicalId\":70759,\"journal\":{\"name\":\"癌症耐药(英文)\",\"volume\":\"8 \",\"pages\":\"42\"},\"PeriodicalIF\":4.6000,\"publicationDate\":\"2025-08-22\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12378350/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"癌症耐药(英文)\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.20517/cdr.2025.124\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2025/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"癌症耐药(英文)","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.20517/cdr.2025.124","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2025/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0
摘要
目的:免疫检查点抑制剂(ICIs)已经改变了癌症治疗;然而,它们对头颈癌(HNC)的疗效仍然有限,只有少数患者实现了持久的反应。因此,了解HNC中ICI耐药性的分子机制至关重要。方法:我们对139例接受ci治疗的HNC患者(MSKCC队列)和502例treatment-naïve HNC患者(TCGA队列)的基因组、转录组学和临床数据进行了综合分析。评估ROS1突变状态、肿瘤突变负荷(TMB)、新抗原负荷、免疫细胞浸润(通过CIBERSORT)和免疫相关基因表达。基因集富集分析(GSEA)用于鉴定失调通路。生存结局采用Kaplan-Meier分析和Cox回归评估,P < 0.05为差异有统计学意义。结果:携带ROS1突变的患者在ICI治疗后表现出明显较差的结果,与ROS1野生型患者相比,中位总生存期更短[OS: 5.0 vs. 11.0个月,风险比(HR) = 3.22, 95%CI: 1.26-8.19, P = 0.011]。多因素分析证实ROS1突变是ci治疗患者生存不良的独立预测因子(HR = 4.78, 95%CI: 1.70 ~ 13.43, P = 0.003)。相比之下,ROS1突变在treatment-naïve TCGA-HNC队列中没有预后意义(P = 0.26),证实了它们作为ICI反应的预测性(非预后)生物标志物的作用。有趣的是,尽管表现出较高的TMB和新抗原水平,ros1突变患者的生存率较低,强调了TMB作为预测性生物标志物的环境依赖性局限性。在机制上,ros1突变肿瘤表现出免疫抑制的肿瘤微环境,其特征是CD8+ T细胞浸润减少,干扰素γ信号减弱,免疫相关基因(CXCL9, CXCL10, IFNG, PD-L1)下调。GSEA显示,在ros1突变肿瘤中MYC通路活性富集,抑制抗原呈递和T细胞活化途径。结论:ROS1突变通过myc介导的转录重编程促进免疫抑制TME,损害抗原呈递和T细胞功能,从而驱动HNC的ICI耐药。将ROS1状态纳入生物标志物面板可以改善患者分层,并指导针对免疫逃避和致癌途径的联合治疗。
ROS1 mutations promote an immunosuppressive tumor microenvironment via MYC to confer immune evasion in head and neck cancer.
Aim: Immune checkpoint inhibitors (ICIs) have transformed cancer therapy; however, their efficacy in head and neck cancer (HNC) remains limited, with only a minority of patients achieving durable responses. Understanding the molecular mechanisms underlying ICI resistance in HNC is therefore crucial. Methods: We conducted an integrative analysis of genomic, transcriptomic, and clinical data from 139 ICI-treated HNC patients (MSKCC cohort) and 502 treatment-naïve HNC cases (TCGA cohort). ROS1 mutation status, tumor mutational burden (TMB), neoantigen load, immune cell infiltration (via CIBERSORT), and immune-related gene expression were evaluated. Gene set enrichment analysis (GSEA) was performed to identify dysregulated pathways. Survival outcomes were assessed using Kaplan-Meier analysis and Cox regression, with statistical significance defined as P < 0.05. Results: Patients harboring ROS1 mutations exhibited significantly poorer outcomes following ICI therapy, with shorter median overall survival [OS: 5.0 vs. 11.0 months, hazard ratio (HR) = 3.22, 95%CI: 1.26-8.19, P = 0.011] compared to ROS1 wild-type counterparts. Multivariate analysis confirmed ROS1 mutation as an independent predictor of poor OS in ICI-treated patients (HR = 4.78, 95%CI: 1.70-13.43, P = 0.003). In contrast, ROS1 mutations showed no prognostic significance in the treatment-naïve TCGA-HNC cohort (P = 0.26), confirming their role as a predictive (not prognostic) biomarker for ICI response. Interestingly, despite exhibiting higher TMB and neoantigen levels, ROS1-mutant patients showed inferior survival, underscoring the context-dependent limitations of TMB as a predictive biomarker. Mechanistically, ROS1-mutant tumors displayed an immunosuppressive tumor microenvironment characterized by diminished CD8+ T cell infiltration, attenuated interferon-γ signaling, and downregulation of immune-related genes (CXCL9, CXCL10, IFNG, PD-L1). GSEA revealed enrichment of MYC pathway activity in ROS1-mutant tumors, which suppressed antigen presentation and T cell activation pathways. Conclusion:ROS1 mutations drive ICI resistance in HNC by promoting an immunosuppressive TME via MYC-mediated transcriptional reprogramming, impairing antigen presentation and T cell function. Incorporating ROS1 status into biomarker panels may improve patient stratification and guide combinatorial therapies targeting both immune evasion and oncogenic pathways.