Anti-galectin-9 therapy synergizes with EGFR inhibition to reprogram the tumor microenvironment and overcome immune evasion.

IF 10.3 1区 医学 Q1 IMMUNOLOGY
Dongli Linghu, Jiaming Song, Liyan Gu, Chengjie Liu, Boning Liu, Riyao Yang, Qihui Chen, Jun Yao, Swathi Priya Manickam, Navaneethan Sundhar, Chih-Yen Tu, Chia-Hung Chen, Zhiguo Liu, Delong Liu, Hsiao-Fan Chen, Shasha Shi, Shunjie Xiong, Mien-Chie Hung, Linlin Sun
{"title":"Anti-galectin-9 therapy synergizes with EGFR inhibition to reprogram the tumor microenvironment and overcome immune evasion.","authors":"Dongli Linghu, Jiaming Song, Liyan Gu, Chengjie Liu, Boning Liu, Riyao Yang, Qihui Chen, Jun Yao, Swathi Priya Manickam, Navaneethan Sundhar, Chih-Yen Tu, Chia-Hung Chen, Zhiguo Liu, Delong Liu, Hsiao-Fan Chen, Shasha Shi, Shunjie Xiong, Mien-Chie Hung, Linlin Sun","doi":"10.1136/jitc-2024-010926","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Despite the remarkable clinical outcomes of epidermal growth factor receptor (EGFR)-targeted therapies in patients with lung cancer, therapeutic resistance eventually develops. This study elucidates the role of galectin-9 (Gal-9), a TIM-3 immune checkpoint ligand, in facilitating tumor immune escape during EGFR tyrosine kinase inhibitor (TKI) therapy, and evaluates the therapeutic potential of combined EGFR-TKI and Gal-9 blockade in preclinical models.</p><p><strong>Methods: </strong>EGFR-TKI-mediated Gal-9 regulation was systematically investigated through multianalysis including RNA-seq transcriptomics, quantitative reverse transcription-PCR, immunoblotting, ELISA, flow cytometry, and immunohistochemical validation across human and murine lung/colorectal cancer cell lines, murine tumor tissues, and paired patient tumor tissues/serum samples. Therapeutic efficacy was evaluated in two syngeneic murine models, with comprehensive immune monitoring of tumor microenvironment (TME), tumor-draining lymph nodes (tdLNs), and splenic compartments. Mechanistic investigations employed CD8<sup>+</sup> T-cell/macrophage depletion strategies (anti-CD8α monoclonal antibodies (mAbs)/PLX-3397), type I interferon (IFN-I) pathway inhibition (anti-IFNAR1 mAbs), and lymph node retention approaches (FTY720 administration).</p><p><strong>Results: </strong>EGFR-TKI treatment significantly induced Gal-9 expression in both tumor cells and host immune cells, particularly myeloid cells. Clinical validation revealed elevated Gal-9 levels in EGFR-TKI-treated patient with lung cancer tumor tissues and serums, correlating with reduced progression-free survival. Mechanistically, EGFR-TKIs triggered DNA damage-potentiated cytosolic double-stranded DNA accumulation and activated tumor-intrinsic STING-IFN-I innate immune pathway that transcriptionally regulated Gal-9 expression. Notably, Gal-9-neutralizing antibodies synergized with EGFR-TKI to markedly inhibit tumor growth in two syngeneic mouse models, including the poorly immunogenic LLC lung tumor model unresponsive to programmed cell death protein-1/programmed death-ligand 1 blockade. The combination therapy remodeled myeloid landscapes toward antigen-presenting phenotypes, promoted dendritic cell accumulation in the tdLN and enhanced CD8<sup>+</sup> T response in the TME. Depleting CD8<sup>+</sup> T cells or macrophages/monocytes abrogated the therapeutic benefits. Blocking the IFN-I pathway attenuated Gal-9 expression and enhanced the antitumor immunity of afatinib in the LLC tumor model.</p><p><strong>Conclusions: </strong>These findings identify Gal-9 upregulation as a key mechanism mediating immune evasion and limiting EGFR-TKI efficacy, providing a promising combinational therapeutic strategy of EGFR-TKI and Gal-9 blockade for the treatment of EGFR-driven cancers.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 7","pages":""},"PeriodicalIF":10.3000,"publicationDate":"2025-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12265805/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal for Immunotherapy of Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1136/jitc-2024-010926","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Despite the remarkable clinical outcomes of epidermal growth factor receptor (EGFR)-targeted therapies in patients with lung cancer, therapeutic resistance eventually develops. This study elucidates the role of galectin-9 (Gal-9), a TIM-3 immune checkpoint ligand, in facilitating tumor immune escape during EGFR tyrosine kinase inhibitor (TKI) therapy, and evaluates the therapeutic potential of combined EGFR-TKI and Gal-9 blockade in preclinical models.

Methods: EGFR-TKI-mediated Gal-9 regulation was systematically investigated through multianalysis including RNA-seq transcriptomics, quantitative reverse transcription-PCR, immunoblotting, ELISA, flow cytometry, and immunohistochemical validation across human and murine lung/colorectal cancer cell lines, murine tumor tissues, and paired patient tumor tissues/serum samples. Therapeutic efficacy was evaluated in two syngeneic murine models, with comprehensive immune monitoring of tumor microenvironment (TME), tumor-draining lymph nodes (tdLNs), and splenic compartments. Mechanistic investigations employed CD8+ T-cell/macrophage depletion strategies (anti-CD8α monoclonal antibodies (mAbs)/PLX-3397), type I interferon (IFN-I) pathway inhibition (anti-IFNAR1 mAbs), and lymph node retention approaches (FTY720 administration).

Results: EGFR-TKI treatment significantly induced Gal-9 expression in both tumor cells and host immune cells, particularly myeloid cells. Clinical validation revealed elevated Gal-9 levels in EGFR-TKI-treated patient with lung cancer tumor tissues and serums, correlating with reduced progression-free survival. Mechanistically, EGFR-TKIs triggered DNA damage-potentiated cytosolic double-stranded DNA accumulation and activated tumor-intrinsic STING-IFN-I innate immune pathway that transcriptionally regulated Gal-9 expression. Notably, Gal-9-neutralizing antibodies synergized with EGFR-TKI to markedly inhibit tumor growth in two syngeneic mouse models, including the poorly immunogenic LLC lung tumor model unresponsive to programmed cell death protein-1/programmed death-ligand 1 blockade. The combination therapy remodeled myeloid landscapes toward antigen-presenting phenotypes, promoted dendritic cell accumulation in the tdLN and enhanced CD8+ T response in the TME. Depleting CD8+ T cells or macrophages/monocytes abrogated the therapeutic benefits. Blocking the IFN-I pathway attenuated Gal-9 expression and enhanced the antitumor immunity of afatinib in the LLC tumor model.

Conclusions: These findings identify Gal-9 upregulation as a key mechanism mediating immune evasion and limiting EGFR-TKI efficacy, providing a promising combinational therapeutic strategy of EGFR-TKI and Gal-9 blockade for the treatment of EGFR-driven cancers.

抗半乳糖凝集素-9治疗与EGFR抑制协同重新编程肿瘤微环境和克服免疫逃避。
背景:尽管表皮生长因子受体(EGFR)靶向治疗肺癌患者的临床效果显著,但治疗耐药性最终会出现。本研究阐明了半乳糖凝集素-9 (Gal-9),一种TIM-3免疫检查点配体,在EGFR酪氨酸激酶抑制剂(TKI)治疗期间促进肿瘤免疫逃逸的作用,并在临床前模型中评估了EGFR-TKI和Gal-9联合阻断的治疗潜力。方法:通过RNA-seq转录组学、定量逆转录pcr、免疫印迹、ELISA、流式细胞术和免疫组化验证等多种分析方法,系统研究egfr - tki介导的Gal-9调控,研究对象包括人和小鼠肺/结直肠癌细胞系、小鼠肿瘤组织和配对患者肿瘤组织/血清样本。通过对肿瘤微环境(TME)、肿瘤引流淋巴结(tdln)和脾室的综合免疫监测,在两种同系性小鼠模型中评估治疗效果。机制研究采用CD8+ t细胞/巨噬细胞消耗策略(抗CD8α单克隆抗体/PLX-3397), I型干扰素(IFN-I)途径抑制(抗ifnar1单克隆抗体)和淋巴结保留方法(FTY720给药)。结果:EGFR-TKI治疗显著诱导肿瘤细胞和宿主免疫细胞,尤其是髓细胞中Gal-9的表达。临床验证显示,egfr - tki治疗的肺癌肿瘤组织和血清中Gal-9水平升高,与无进展生存期降低相关。在机制上,EGFR-TKIs触发DNA损伤增强的细胞质双链DNA积累,激活肿瘤固有的STING-IFN-I先天免疫途径,转录调节Gal-9的表达。值得注意的是,gal -9中和抗体与EGFR-TKI协同作用,在两种同基因小鼠模型中显著抑制肿瘤生长,包括对程序性细胞死亡蛋白-1/程序性死亡配体1阻断无反应的低免疫原性LLC肺肿瘤模型。联合治疗重塑了抗原呈递表型的骨髓景观,促进了tdLN中的树突状细胞积聚,增强了TME中的CD8+ T反应。消耗CD8+ T细胞或巨噬细胞/单核细胞会使治疗效果消失。在LLC肿瘤模型中,阻断IFN-I通路可降低Gal-9的表达,增强阿法替尼的抗肿瘤免疫。结论:这些发现确定了Gal-9上调是介导免疫逃避和限制EGFR-TKI疗效的关键机制,为EGFR-TKI和Gal-9阻断治疗egfr驱动的癌症提供了一种有希望的联合治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Journal for Immunotherapy of Cancer
Journal for Immunotherapy of Cancer Biochemistry, Genetics and Molecular Biology-Molecular Medicine
CiteScore
17.70
自引率
4.60%
发文量
522
审稿时长
18 weeks
期刊介绍: The Journal for ImmunoTherapy of Cancer (JITC) is a peer-reviewed publication that promotes scientific exchange and deepens knowledge in the constantly evolving fields of tumor immunology and cancer immunotherapy. With an open access format, JITC encourages widespread access to its findings. The journal covers a wide range of topics, spanning from basic science to translational and clinical research. Key areas of interest include tumor-host interactions, the intricate tumor microenvironment, animal models, the identification of predictive and prognostic immune biomarkers, groundbreaking pharmaceutical and cellular therapies, innovative vaccines, combination immune-based treatments, and the study of immune-related toxicity.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信