Transitional CXCL14+ cancer-associated fibroblasts enhance tumour metastasis and confer resistance to EGFR-TKIs, revealing therapeutic vulnerability to filgotinib in lung adenocarcinoma

Weijiao Xu , Haitang Yang , Ke Xu , Anshun Zhu , Sean R. R. Hall , Yunxuan Jia , Baicheng Zhao , Enshuo Zhang , Gang Liu , Jianlin Xu , Thomas M. Marti , Ren-Wang Peng , Patrick Dorn , Yongliang Niu , Xufeng Pan , Yajuan Zhang , Feng Yao

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70281

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70281 DOI: 10.1002/ctm2.70281
RESEARCH ARTICLE

Transitional CXCL14+ cancer-associated fibroblasts enhance tumour metastasis and confer resistance to EGFR-TKIs, revealing therapeutic vulnerability to filgotinib in lung adenocarcinoma

Author information +
History +
PDF

Abstract

Background: The heterogeneity of cancer-associated fibroblasts (CAFs) has become a crucial focus in understanding cancer biology and treatment response, revealing distinct subpopulations with specific roles in tumor pathobiology. CAFs have also been shown to promote resistance in lung cancer cells to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs). However, the specific CAF subsets responsible for driving tumor advancement and resistance to EGFR-TKIs in lung adenocarcinoma (LUAD) remain poorly understood.

Methods: We integrate multiple scRNA-seq datasets to identify cell subclusters most relevant to tumor stage, patient survival, and EGFR–TKIs response. Additionally, in vitro and in vivo experiments, clinical tissue sample immunohistochemistry and patient plasma ELISA experiments are performed to validate key findings in independent LUAD cohorts.

Results: By analyzing multiple scRNA-seq and spatial transcriptomic datasets, we identified a unique subset of CXCL14+ myofibroblastic CAFs (myCAFs), emerging during the early differentiation phase of pan-cancer invasiveness-associated THBS2+ POSTN+ COL11A1+ myCAFs. Notably, plasma levels of CXCL14 in LUAD patients correlate significantly with tumor stage. Mechanistically, this subset enhances tumor aggressiveness through epithelial-to-mesenchymal transition and angiogenesis. Among standard treatment regimens, transitional CXCL14+ myCAFs specifically confer resistance to EGFR-TKIs, while showing no significant impact on chemotherapy or immunotherapy outcomes. Through a pharmacological screen of FDA-approved drugs, we identified Filgotinib as an effective agent to counteract the EGFR-TKIs resistance conferred by this CAF subset.

Conclusions: In summary, our study highlights the role of the differentiated stage from transitional CXCL14+ myCAFs to invasiveness-associated myCAFs in driving tumor progression and therapy resistance in LUAD, positioning Filgotinib as a promising targeted therapy for this process. These insights may enhance patient stratification and inform precision treatment strategies in LUAD.

Keywords

cancer-associated fibroblasts / EGFR-TKIs / heterogeneity / lung adenocarcinoma / metastasis / survival

Cite this article

Download citation ▾
Weijiao Xu, Haitang Yang, Ke Xu, Anshun Zhu, Sean R. R. Hall, Yunxuan Jia, Baicheng Zhao, Enshuo Zhang, Gang Liu, Jianlin Xu, Thomas M. Marti, Ren-Wang Peng, Patrick Dorn, Yongliang Niu, Xufeng Pan, Yajuan Zhang, Feng Yao. Transitional CXCL14+ cancer-associated fibroblasts enhance tumour metastasis and confer resistance to EGFR-TKIs, revealing therapeutic vulnerability to filgotinib in lung adenocarcinoma. Clinical and Translational Medicine, 2025, 15(4): e70281 DOI:10.1002/ctm2.70281

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024; 74(1): 12-49.

[2]

Gridelli C, Rossi A, Carbone DP, et al. Non-small-cell lung cancer. Nat Rev Dis Primers. 2015; 1: 15009.

[3]

Neophytou CM, Panagi M, Stylianopoulos T, et al. The role of tumor microenvironment in cancer metastasis: molecular mechanisms and therapeutic opportunities. Cancers (Basel). 2021; 13(9): 2053.

[4]

De Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023; 41(3): 374-403.

[5]

Chhabra Y, Weeraratna AT. Fibroblasts in cancer: unity in heterogeneity. Cell. 2023; 186(8): 1580-1609.

[6]

Lavie D, Ben-Shmuel A, Erez N, et al. Cancer-associated fibroblasts in the single-cell era. Nat Cancer. 2022; 3(7): 793-807.

[7]

Davidson S, Coles M, Thomas T, et al. Fibroblasts as immune regulators in infection, inflammation and cancer. Nat Rev Immunol. 2021; 21(11): 704-717.

[8]

Ohlund D, Handly-Santana A, Biffi G, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 2017; 214(3): 579-596.

[9]

Elyada E, Bolisetty M, Laise P, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019; 9(8): 1102-1123.

[10]

Biffi G, Tuveson DA. Diversity and biology of cancer-associated fibroblasts. Physiol Rev. 2021; 101(1): 147-176.

[11]

Chen Y, Mcandrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021; 18(12): 792-804.

[12]

Yano S, Wang W, Li Q, et al. Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor-activating mutations. Cancer Res. 2008; 68(22): 9479-9487.

[13]

Dai S, Liu Y, Liu Z, et al. Cancer-associated fibroblasts mediate resistance to anti-EGFR therapies in cancer. Pharmacol Res. 2024; 206: 107304.

[14]

Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018; 553(7689): 446-454.

[15]

Ramalingam SS, Yang JC, Lee CK, et al. Osimertinib as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer. J Clin Oncol. 2018; 36(9): 841-849.

[16]

Yuan M, Huang LL, Chen JH, et al. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer. Signal Transduct Target Ther. 2019; 4: 61.

[17]

Grigg C, Rizvi NA. PD-L1 biomarker testing for non-small cell lung cancer: truth or fiction? J Immunother Cancer. 2016; 4: 48.

[18]

Martinez P, Peters S, Stammers T, et al. Immunotherapy for the first-line treatment of patients with metastatic non-small cell lung cancer. Clin Cancer Res. 2019; 25(9): 2691-2698.

[19]

Kim N, Kim HK, Lee K, et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat Commun. 2020; 11(1): 2285.

[20]

Xing X, Yang F, Huang Q, et al. Decoding the multicellular ecosystem of lung adenocarcinoma manifested as pulmonary subsolid nodules by single-cell RNA sequencing. Sci Adv. 2021; 7(5): eabd9738.

[21]

Laughney AM, Hu J, Campbell NR, et al. Regenerative lineages and immune-mediated pruning in lung cancer metastasis. Nat Med. 2020; 26(2): 259-269.

[22]

Wu F, FAN J, He Y, et al. Single-cell profiling of tumor heterogeneity and the microenvironment in advanced non-small cell lung cancer. Nat Commun. 2021; 12(1): 2540.

[23]

Zappia L, Oshlack A. Clustering trees: a visualization for evaluating clusterings at multiple resolutions. Gigascience. 2018; 7(7): giy083.

[24]

Cancer Genome Atlas Research N. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014; 511(7511): 543-550.

[25]

Landi MT, Dracheva T, Rotunno M, et al. Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival. PLoS One. 2008; 3(2): e1651.

[26]

Rousseaux S, Debernardi A, Jacquiau B, et al. Ectopic activation of germline and placental genes identifies aggressive metastasis-prone lung cancers. Sci Transl Med. 2013; 5(186): 186ra66.

[27]

Okayama H, Kohno T, Ishii Y, et al. Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 2012; 72(1): 100-111.

[28]

Selamat SA, Chung BS, Girard L, et al. Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res. 2012; 22(7): 1197-1211.

[29]

Robles AI, Arai E, Mathe EA, et al. An integrated prognostic classifier for stage I lung adenocarcinoma based on mRNA, microRNA, and DNA methylation biomarkers. J Thorac Oncol. 2015; 10(7): 1037-1048.

[30]

Beer DG, Kardia SL, Huang CC, et al. Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat Med. 2002; 8(8): 816-824.

[31]

Zhu K, Cai L, Cui C, et al. Single-cell analysis reveals the pan-cancer invasiveness-associated transition of adipose-derived stromal cells into COL11A1-expressing cancer-associated fibroblasts. PLoS Comput Biol. 2021; 17(7): e1009228.

[32]

Schabath MB, Welsh EA, Fulp WJ, et al. Differential association of STK11 and TP53 with KRAS mutation-associated gene expression, proliferation and immune surveillance in lung adenocarcinoma. Oncogene. 2016; 35(24): 3209-3216.

[33]

Zhu J, Fan Y, Xiong Y, et al. Delineating the dynamic evolution from preneoplasia to invasive lung adenocarcinoma by integrating single-cell RNA sequencing and spatial transcriptomics. Exp Mol Med. 2022; 54(11): 2060-2076.

[34]

Maynard A, Mccoach CE, Rotow JK, et al. Therapy-induced evolution of human lung cancer revealed by single-cell RNA sequencing. Cell. 2020; 182(5): 1232-1251.e22.

[35]

Qureshy Z, Johnson DE, Grandis JR. Targeting the JAK/STAT pathway in solid tumors. J Cancer Metastasis Treat. 2020; 6: 27.

[36]

Genovese MC, Kalunian K, Gottenberg JE, et al. Effect of filgotinib vs placebo on clinical response in patients with moderate to severe rheumatoid arthritis refractory to disease-modifying antirheumatic drug therapy: the FINCH 2 randomized clinical trial. JAMA. 2019; 322(4): 315-325.

[37]

Tarrant JM, Galien R, Li W, et al. Filgotinib, a JAK1 inhibitor, modulates disease-related biomarkers in rheumatoid arthritis: results from two randomized, controlled phase 2b trials. Rheumatol Ther. 2020; 7(1): 173-190.

[38]

Van Rompaey L, Galien R, van der Aar EM, et al. Preclinical characterization of GLPG0634, a selective inhibitor of JAK1, for the treatment of inflammatory diseases. J Immunol. 2013; 191(7): 3568-3577.

[39]

Barrett RL, Pure E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. Elife. 2020; 9: e57243.

[40]

Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016; 16(9): 582-598.

[41]

Ohlund D, Elyada E, Tuveson D. Fibroblast heterogeneity in the cancer wound. J Exp Med. 2014; 211(8): 1503-1523.

[42]

Sahai E, Astsaturov I, Cukierman E, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020; 20(3): 174-186.

[43]

Mao X, Xu J, Wang W, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021; 20(1): 131.

[44]

Hromas R, Broxmeyer HE, Kim C, et al. Cloning of BRAK, a novel divergent CXC chemokine preferentially expressed in normal versus malignant cells. Biochem Biophys Res Commun. 1999; 255(3): 703-706.

[45]

Gowhari S, Haleem A-Q, Markov A, et al. Chemokine CXCL14; a double-edged sword in cancer development. Int Immunopharmacol. 2021; 97: 107681.

[46]

Augsten M, Hägglöf C, Olsson E, et al. CXCL14 is an autocrine growth factor for fibroblasts and acts as a multi-modal stimulator of prostate tumor growth. Proc Natl Acad Sci U S A. 2009; 106(9): 3414-3419.

[47]

Xu Y, Deng C, Chen H, et al. Osteosarcoma cells secrete CXCL14 that activates integrin α11β1 on fibroblasts to form a lung metastatic niche. Cancer Res. 2024; 84(7): 994-1012.

[48]

Chang TM, Chiang YC, Lee CW, et al. CXCL14 promotes metastasis of non-small cell lung cancer through ACKR2-depended signaling pathway. Int J Biol Sci. 2023; 19(5): 1455-1470.

[49]

Cai L, Kolonin MG, Anastassiou D. The fibro-adipogenic progenitor APOD+DCN+LUM+ cell population in aggressive carcinomas. Cancer Metastasis Rev. 2024; 43(3): 977-980.

[50]

Yan Y, Sun D, Hu J, et al. Multi-omic profiling highlights factors associated with resistance to immuno-chemotherapy in non-small-cell lung cancer. Nat Genet. 2025; 57(1): 126-139.

[51]

Moffitt RA, Marayati R, Flate EL, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015; 47(10): 1168-1178.

[52]

Hu Y, Taylor-Harding B, Raz Y, et al. Are epithelial ovarian cancers of the mesenchymal subtype actually intraperitoneal metastases to the ovary? Front Cell Dev Biol. 2020; 8: 647.

[53]

Wu SG, Shih JY. Management of acquired resistance to EGFR TKI-targeted therapy in advanced non-small cell lung cancer. Mol Cancer. 2018; 17(1): 38.

[54]

Yi Y, Zeng S, Wang Z, et al. Cancer-associated fibroblasts promote epithelial-mesenchymal transition and EGFR-TKI resistance of non-small cell lung cancers via HGF/IGF-1/ANXA2 signaling. Biochim Biophys Acta Mol Basis Dis. 2018; 1864(3): 793-803.

[55]

Akli A, Girard N, Fallet V, et al. Histomolecular resistance mechanisms to first-line osimertinib in EGFR-mutated advanced non-small cell lung cancer: a multicentric retrospective French study. Target Oncol. 2022; 17(6): 675-682.

[56]

Oxnard GR, Hu Y, Mileham KF, et al. Assessment of resistance mechanisms and clinical implications in patients with EGFR T790M-positive lung cancer and acquired resistance to osimertinib. JAMA Oncol. 2018; 4(11): 1527-1534.

[57]

Chmielecki J, Gray JE, Cheng Y, et al. Candidate mechanisms of acquired resistance to first-line osimertinib in EGFR-mutated advanced non-small cell lung cancer. Nat Commun. 2023; 14(1): 1070.

[58]

Schoenfeld AJ, Chan JM, Kubota D, et al. Tumor analyses reveal squamous transformation and off-target alterations as early resistance mechanisms to first-line osimertinib in EGFR-mutant lung cancer. Clin Cancer Res. 2020; 26(11): 2654-2663.

[59]

Piotrowska Z, Isozaki H, Lennerz JK, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov. 2018; 8(12): 1529-1539.

[60]

Le X, Puri S, Negrao MV, et al. Landscape of EGFR-dependent and -independent resistance mechanisms to osimertinib and continuation therapy beyond progression in EGFR-mutant NSCLC. Clin Cancer Res. 2018; 24(24): 6195-6203.

[61]

Kim DM, Kim MJ, Moon JH, et al. Inhibition of JAK1/2 can overcome EGFR-TKI resistance in human NSCLC. Biochem Biophys Res Commun. 2020; 527(1): 305-310.

[62]

Kim SM, Kwon OJ, Hong YK, et al. Activation of IL-6R/JAK1/STAT3 signaling induces de novo resistance to irreversible EGFR inhibitors in non-small cell lung cancer with T790M resistance mutation. Mol Cancer Ther. 2012; 11(10): 2254-2264.

[63]

Zak J, Pratumchai I, Marro BS, et al. JAK inhibition enhances checkpoint blockade immunotherapy in patients with Hodgkin lymphoma. Science. 2024; 384(6702): eade8520.

[64]

Mathew D, Marmarelis ME, Foley C, et al. Combined JAK inhibition and PD-1 immunotherapy for non-small cell lung cancer patients. Science. 2024; 384(6702): eadf1329.

[65]

Shi M, Chen P, Cui B, et al. Benmelstobart plus anlotinib in patients with EGFR-positive advanced NSCLC after failure of EGFR TKIs therapy: a phase I/II study. Signal Transduct Target Ther. 2024; 9(1): 283.

[66]

Blumenschein G, Chandler J, Garon EB, et al. PS01.59: CheckMate 370: a master protocol of phase 1/2 studies of nivolumab as maintenance or first-line ± standard-of-care therapies in advanced NSCLC. J Thorac Oncol. 2016; 11(11): S307.

[67]

Yang JC, Gadgeel SM, Sequist LV, et al. Pembrolizumab in combination with erlotinib or gefitinib as first-line therapy for advanced NSCLC with sensitizing EGFR mutation. J Thorac Oncol. 2019; 14(3): 553-559.

[68]

Yang H, Sun B, Xu K, et al. Pharmaco-transcriptomic correlation analysis reveals novel responsive signatures to HDAC inhibitors and identifies Dasatinib as a synergistic interactor in small-cell lung cancer. EBioMedicine. 2021; 69: 103457.

[69]

Yang H, Liang SQ, Xu D, et al. HSP90/AXL/eIF4E-regulated unfolded protein response as an acquired vulnerability in drug-resistant KRAS-mutant lung cancer. Oncogenesis. 2019; 8(9): 45.

[70]

Yang H, Zhao L, Gao Y, et al. Pharmacotranscriptomic analysis reveals novel drugs and gene networks regulating ferroptosis in cancer. Cancers (Basel). 2020; 12(11): 3273.

[71]

Zheng S, Wang W, Aldahdooh J, et al. SynergyFinder plus: toward better interpretation and annotation of drug combination screening datasets. Genomics Proteomics Bioinformatics. 2022; 20(3): 587-596.

[72]

Russo A, Franchina T, Ricciardi GRR, et al. Third generation EGFR TKIs in EGFR-mutated NSCLC: where are we now and where are we going. Crit Rev Oncol Hematol. 2017; 117: 38-47.

[73]

Yang H, Sun B, Ma W, et al. Multi-scale characterization of tumor-draining lymph nodes in resectable lung cancer treated with neoadjuvant immune checkpoint inhibitors. EBioMedicine. 2022; 84: 104265.

[74]

Cho JW, Hong MH, Ha SJ, et al. Genome-wide identification of differentially methylated promoters and enhancers associated with response to anti-PD-1 therapy in non-small cell lung cancer. Exp Mol Med. 2020; 52(9): 1550-1563.

[75]

Jung H, Kim HS, Kim JY, et al. DNA methylation loss promotes immune evasion of tumours with high mutation and copy number load. Nat Commun. 2019; 10(1): 4278.

[76]

Hwang S, Kwon AY, Jeong JY, et al. Immune gene signatures for predicting durable clinical benefit of anti-PD-1 immunotherapy in patients with non-small cell lung cancer. Sci Rep. 2020; 10(1): 643.

[77]

Prat A, Navarro A, Pare L, et al. Immune-related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Cancer Res. 2017; 77(13): 3540-3550.

[78]

Hatzis C, Pusztai L, Valero V, et al. A genomic predictor of response and survival following taxane-anthracycline chemotherapy for invasive breast cancer. JAMA. 2011; 305(18): 1873-1881.

[79]

Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019; 177(7): 1888-1902.e21.

[80]

Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019; 20(1): 296.

[81]

Jin S, Guerrero-Juarez CF, Zhang L, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun. 2021; 12(1): 1088.

[82]

Andreatta M, Carmona SJ. UCell: robust and scalable single-cell gene signature scoring. Comput Struct Biotechnol J. 2021; 19: 3796-3798.

[83]

Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013; 14: 7.

[84]

Wu T, Hu E, Xu S, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb). 2021; 2(3): 100141.

[85]

Liberzon A, Birger C, Thorvaldsdottir H, et al. The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 2015; 1(6): 417-425.

[86]

Kim D, Paggi JM, Park C, et al. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019; 37(8): 907-915.

[87]

Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15(12): 550.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

8

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/