Abstract
Although immune checkpoint blockade (ICB) therapies have shifted the treatment paradigm for non-small-cell lung cancer (NSCLC), many patients remain resistant. Here we characterize the tumor cell states and spatial cellular compositions of the NSCLC tumor microenvironment (TME) by analyzing single-cell transcriptomes of 232,080 cells and spatially resolved transcriptomes of tumors from 19 patients before and after ICB–chemotherapy. We find that tumor cells and secreted phosphoprotein 1-positive macrophages interact with collagen type XI alpha 1 chain-positive cancer-associated fibroblasts to stimulate the deposition and entanglement of collagen fibers at tumor boundaries, obstructing T cell infiltration and leading to poor prognosis. We also reveal distinct states of tertiary lymphoid structures (TLSs) in the TME. Activated TLSs are associated with improved prognosis, whereas a hypoxic microenvironment appears to suppress TLS development and is associated with poor prognosis. Our study provides novel insights into different cellular and molecular components corresponding to NSCLC ICB–chemotherapeutic responsiveness, which will benefit future individualized immuno-chemotherapy.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout







Similar content being viewed by others
Data availability
Raw scRNA-seq and spatial transcriptomics data have been deposited to the Genome Sequence Archive of the BIG Data Center at the Beijing Institute of Genomics, Chinese Academy of Science, under accession number HRA002509 (accessible at http://bigd.big.ac.cn/gsa-human). Processed bulk RNA-seq, scRNA-seq and spatial transcriptomics data are available from Zenodo (accessible at https://zenodo.org/records/8227624)76. The bulk RNA-seq data of the independent NSCLC cohort6 were acquired from the Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo) with the accession GSE207422. The bulk RNA-seq data of the Stand Up To Cancer–Mark Foundation for Cancer Research cohort31 are available from Zenodo (accessible at https://zenodo.org/records/7849582)77. The bulk RNA-seq data of the OAK and POPLAR cohorts30 were acquired from the European Genome-Phenome Archive (https://ega-archive.org) with the accession EGAS00001005013. The bulk RNA-seq data of the melanoma cohort32 were acquired from the Gene Expression Omnibus with the accession GSE78220.
Code availability
The code for identifying the TLSs in spatial transcriptomics slides can be found at https://github.com/wanglabtongji/Scanner. Code for inferring the cell type distribution in spatial transcriptomics data can be found at https://github.com/wanglabtongji/STRIDE.
References
Brahmer, J. et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med. 373, 123–135 (2015).
Socinski, M. A. et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 378, 2288–2301 (2018).
Bian, D. L. et al. Neoadjuvant Afatinib for stage III EGFR-mutant non-small cell lung cancer: a phase II study. Nat. Commun. 14, 4655 (2023).
Lu, S. et al. Perioperative toripalimab plus chemotherapy for patients with resectable non-small cell lung cancer: the Neotorch Randomized Clinical Trial. J. Am. Med. Assoc. 331, 201–211 (2024).
Xia, H. R. et al. Neoadjuvant camrelizumab (an anti-PD-1 antibody) plus chemotherapy or apatinib (a VEGFR-2 inhibitor) for initially unresectable stage II-III non-small-cell lung cancer: a multicentre, two-arm, phase 2 exploratory study. Signal Transduct. Target. Ther. 9, 145 (2024).
Zhu, X. et al. Safety and effectiveness of neoadjuvant PD-1 inhibitor (toripalimab) plus chemotherapy in stage II-III NSCLC (LungMate 002): an open-label, single-arm, phase 2 trial. BMC Med. 20, 493 (2022).
Jia, X. H. et al. Efficacy and safety of neoadjuvant immunotherapy in resectable nonsmall cell lung cancer: a meta-analysis. Lung Cancer 147, 143–153 (2020).
Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).
Caushi, J. X. et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 596, 126–132 (2021).
Liu, B. L. et al. Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat. Cancer 3, 108–121 (2022).
Sautes-Fridman, C. et al. Tertiary lymphoid structures in cancers: prognostic value, regulation, and manipulation for therapeutic intervention. Front. Immunol. 7, 407 (2016).
Sautes-Fridman, C., Petitprez, F., Calderaro, J. & Fridman, W. H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 19, 307–325 (2019).
Petitprez, F. et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 577, 556–560 (2020).
Vanhersecke, L. et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat. Cancer 2, 794–802 (2021).
Travis, W. D. et al. IASLC multidisciplinary recommendations for pathologic assessment of lung cancer resection specimens after neoadjuvant therapy. J. Thorac. Oncol. 15, 709–740 (2020).
William, W. N. et al. Computed tomography RECIST assessment of histopathologic response and prediction of survival in patients with resectable non-small-cell lung cancer after neoadjuvant chemotherapy. J. Thorac. Oncol. 8, 222–228 (2013).
Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).
Barkley, D. et al. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat. Genet. 54, 1192–1201 (2022).
Neftel, C. et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell 178, 835–849.e21 (2019).
Puram, S. V. et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171, 1611–1624.e24 (2017).
Tirosh, I. et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539, 309–313 (2016).
Fu, T. et al. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J. Hematol. Oncol. 14, 98 (2021).
Sun, D. Q., Liu, Z. Y., Li, T. W., Wu, Q. & Wang, C. F. STRIDE: accurately decomposing and integrating spatial transcriptomics using single-cell RNA sequencing. Nucleic Acids Res. 50, e42 (2022).
Ohlund, D. et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 214, 579–596 (2017).
Krishnamurty, A. T. et al. LRRC15+ myofibroblasts dictate the stromal setpoint to suppress tumour immunity. Nature 611, 148–154 (2022).
Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).
Guido, C. et al. Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: connecting TGF-β signaling with “Warburg-like” cancer metabolism and l-lactate production. Cell Cycle 11, 3019–3035 (2012).
Weinstein, J. N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45, 1113–1120 (2013).
Sun, X. J. et al. Tumour DDR1 promotes collagen fibre alignment to instigate immune exclusion. Nature 599, 673–678 (2021).
Patil, N. S. et al. Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. Cancer Cell 40, 289–300.e4 (2022).
Ravi, A. et al. Genomic and transcriptomic analysis of checkpoint blockade response in advanced non-small cell lung cancer. Nat. Genet. 55, 807–819 (2023).
Hugo, W. et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).
Qi, J. J. et al. Single-cell and spatial analysis reveal interaction of FAP+ fibroblasts and SPP1+ macrophages in colorectal cancer. Nat. Commun. 13, 1742 (2022).
Sathe, A. et al. Colorectal cancer metastases in the liver establish immunosuppressive spatial networking between tumor-associated SPP1+ macrophages and fibroblasts. Clin. Cancer Res. 29, 244–260 (2023).
Chen, S. Q. et al. β-catenin-controlled tubular cell-derived exosomes play a key role in fibroblast activation via the OPN–CD44 axis. J. Extracell. Vesicles 11, e12203 (2022).
Silina, K. et al. Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res. 78, 1308–1320 (2018).
Posch, F. et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. OncoImmunology 7, e1378844 (2018).
Baird, N. A., Turnbull, D. W. & Johnson, E. A. Induction of the heat shock pathway during hypoxia requires regulation of heat shock factor by hypoxia-inducible factor-1. J. Biol. Chem. 281, 38675–38681 (2006).
Facciabene, A. et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 475, 226–230 (2011).
Ren, L. et al. Hypoxia-induced CCL28 promotes recruitment of regulatory T cells and tumor growth in liver cancer. Oncotarget 7, 75763–75773 (2016).
Westendorf, A. M. et al. Hypoxia enhances immunosuppression by inhibiting CD4+ effector T cell function and promoting Treg activity. Cell. Physiol. Biochem. 41, 1271–1284 (2017).
Da Silva-Diz, V., Lorenzo-Sanz, L., Bernat-Peguera, A., Lopez-Cerda, M. & Munoz, P. Cancer cell plasticity: impact on tumor progression and therapy response. Semin. Cancer Biol. 53, 48–58 (2018).
Qin, S. et al. Emerging role of tumor cell plasticity in modifying therapeutic response. Signal Transduct. Target. Ther. 5, 1995–2030 (2020).
Fan, J. et al. Multiomic analysis of cervical squamous cell carcinoma identifies cellular ecosystems with biological and clinical relevance. Nat. Genet. 55, 2175–2188 (2023).
Singh, A. et al. NRF2 activation promotes aggressive lung cancer and associates with poor clinical outcomes. Clin. Cancer Res. 27, 877–888 (2021).
Jeong, Y. et al. Role of KEAP1/NFE2L2 mutations in the chemotherapeutic response of patients with non-small cell lung cancer. Clin. Cancer Res. 26, 274–281 (2020).
Krail, E. B. et al. KEAP1 loss modulates sensitivity to kinase targeted therapy in lung cancer. eLife 6, e18970 (2017).
Duan, J. et al. Tumor-immune microenvironment and NRF2 associate with clinical efficacy of PD-1 blockade combined with chemotherapy in lung squamous cell carcinoma. Cell Rep. Med. 4, 101302 (2023).
Sugimoto, H., Mundel, T. M., Kieran, M. W. & Kalluri, R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol. Ther. 5, 1640–1646 (2006).
Ozdemir, B. C. et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25, 719–734 (2014).
Ohlund, D., Elyada, E. & Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 211, 1503–1523 (2014).
Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 (2016).
Dominguez, C. X. et al. Single-cell RNA sequencing reveals stromal evolution into LRRC15+ myofibroblasts as a determinant of patient response to cancer immunotherapy. Cancer Discov. 10, 232–253 (2020).
Kieffer, Y. et al. Single-cell analysis reveals fibroblast clusters linked to immunotherapy resistance in cancer. Cancer Discov. 10, 1330–1351 (2020).
Chauhan, V. P. et al. Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy. Proc. Natl Acad. Sci. USA 116, 10674–10680 (2019).
Lu, C. W. et al. WDR5–H3K4me3 epigenetic axis regulates OPN expression to compensate PD-L1 function to promote pancreatic cancer immune escape. J. Immunother. Cancer 9, e002624 (2021).
Zhu, Y. et al. Disruption of tumour-associated macrophage trafficking by the osteopontin-induced colony-stimulating factor-1 signalling sensitises hepatocellular carcinoma to anti-PD-L1 blockade. Gut 68, 1653–1666 (2019).
Cabrita, R. et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 577, 561–565 (2020).
Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).
Fridman, W. H. et al. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat. Rev. Clin. Oncol. 19, 441–457 (2022).
Merritt, C. R. et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol. 38, 586–599 (2020).
Zhao, Y., Wong, L. & Goh, W. W. B. How to do quantile normalization correctly for gene expression data analyses. Sci. Rep. 10, 15534 (2020).
Chen, Y. et al. Deep autoencoder for interpretable tissue-adaptive deconvolution and cell-type-specific gene analysis. Nat. Commun. 13, 6735 (2022).
Hao, Y. H. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).
Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).
Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).
Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).
Haenzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).
Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).
Cabello-Aguilar, S. et al. SingleCellSignalR: inference of intercellular networks from single-cell transcriptomics. Nucleic Acids Res. 48, e55 (2020).
Cheng, S. J. et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell 184, 792–809.e23 (2021).
Newman, A. M. et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 37, 773–782 (2019).
Luca, B. A. et al. Atlas of clinically distinct cell states and ecosystems across human solid tumors. Cell 184, 5482–5496.e28 (2021).
Qiu, X. J. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).
Germain, C., Gnjatic, S. & Dieu-Nosjean, M. C. Tertiary lymphoid structure-associated B cells are key players in anti-tumor immunity. Front. Immunol. 6, 67 (2015).
Yan, Y. Multi-omic profiling highlights factors associated with resistance to immuno-chemotherapy in non-small cell lung cancer. Zenodo https://doi.org/10.5281/zenodo.13901289 (2024).
Holton, M., Arniella, M., Ravi, A. & Getz, G. Genomic and transcriptomic analysis of checkpoint blockade response in advanced non-small cell lung cancer. Zenodo https://doi.org/10.5281/zenodo.7849582 (2023).
Acknowledgements
This work was supported by the National Natural Science Foundation of China (82125001 (P.Z.), 62088101 (C.W.), 82201948 (L.Z.), 32170660 (C.W.) and 82030035 (Y.E.S.)), National Key R&D Program of China (grant 2022YFA1106000 (C.W.)), Clinical Research Plan of Shanghai Hospital Development Center (grant SHDC2020CR2020B (P.Z.)), Innovation Program of the Shanghai Municipal Education Commission (grant 2023ZKZD33 (P.Z.)), Shanghai Pulmonary Hospital (FKCX1904 (P.Z.), FKYQ2308 (L.Z.) and FKLY20004 (P.Z.)), Shanghai Pilot Program for Basic Research (C.W.), Shanghai Municipal Science and Technology Major Project (2021SHZDZX0100 (C.W.)), Peak Disciplines (Type IV) of Institutions of Higher Learning in Shanghai (C.W. and Y.E.S.) and Fundamental Research Funds for the Central Universities (22120240435 (C.W.)).
Author information
Authors and Affiliations
Contributions
P.Z., C.W., Y.E.S., L.Z. and A.Z. conceived of and designed the study. H.Y., Y.X., Z.H., H.X., X.Z., D.B., F.S., Y.C., L.H., C.W., A.Z. and L.Z. developed and performed the experiments or collected data. Y.Y., D.S., J.H., L.S. and C.W. designed and performed the computation and statistical analyses. Y.Y., D.S., J.H., O.R.F., H.H., L.Z., Y.E.S., C.W. and P.Z. wrote, reviewed and edited the manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Genetics thanks Sydney Shaffer and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Characterization of immunotherapy-treated NSCLC by combined scRNA-seq and spatial transcriptomic analysis.
a, Dot-heat plot showing marker genes for each major lineage. The color gradient represents the expression level, and the diameter represents the percentage of cells. b, Examples of inferred CNV profiles of malignant cells and normal epithelial cells, with endothelium and CD31− stromal cells as references. Left panel: lung squamous cell carcinoma (LUSC). Right panel: lung adenocarcinoma (LUAD). c, Dot-heat plot showing the markers of adeno, squamous and neuroendocrine tumors in P08 and P17.d, UMAP plot of all epithelial cells colored by subpopulations. e, Boxplot showing the CNV score of each epithelium subpopulation. Box limits denote the first and third quartiles with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box. f, Dot-heat plot showing the marker genes for each normal epithelial cluster. The color gradient represents the scaled expression level, and the diameter represents the percentage of cells. g, Bar plot showing the percentage of major lineages for each sample. Related to Fig. 1.
Extended Data Fig. 2 Characterization of immunotherapy-treated NSCLC by combined scRNA-seq and spatial transcriptomic analysis.
a, Dot-heat plot showing the marker genes for each T cluster. The color gradient represents the scaled expression level, and the diameter represents the percentage of cells. b, Dot-heat plot showing the marker genes for each B cluster. The color gradient represents the scaled expression level, and the diameter represents the percentage of cells. c, Dot-heat plot showing the marker genes for each Mono/Macro cluster. The color gradient represents the scaled expression level, and the diameter represents the percentage of cells. d, Dot-heat plot showing the marker genes for each CD31− stromal cluster. The color gradient represents the scaled expression level, and the diameter represents the percentage of cells. e, Dot-heat plot showing the marker genes for each DC cluster. The color gradient represents the scaled expression level, and the diameter represents the percentage of cells. f, Dot-heat plot showing the marker genes for each endothelial cluster. The color gradient represents the scaled expression level, and the diameter represents the percentage of cells. Related to Fig. 1.
Extended Data Fig. 3 Spatial mapping of the cellular context in NSCLC after ICB-chemotherapy.
a, Scaled average cell-type compositions within each cell-type context. Related to Fig. 3.
Extended Data Fig. 4 Single-cell and spatial transcriptomic analyses of fibroblast subsets after ICB-chemotherapy.
a, Violin plots showing iCAF and myCAF signature scores in COL11A1+ CAFs (n = 2359) and ADH1B+ CAFs (n = 956). Box limits denote the first and third quartiles with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box. b, Heatmap showing differentially enriched Hallmark and KEGG metabolic pathways in CD31− stromal subsets. c, Heatmap showing potential ligands driving the phenotype of COL11A1+ CAF cells. d, Boxplots showing the average expression of COL11A1 in COL11A1+ CAF cells from pre-treatment responders (PreR, n = 3), pre-treatment non-responders (PreNR, n = 4), post-treatment responders (PostR, n = 5) and post-treatment non-responders (PostNR, n = 12). The center line indicates the median, and the lower and upper hinges represent the 25th and 75th percentiles, respectively. Whiskers denote 1.5× interquartile range. A two-sided t test was used to determine the statistical significance. e, Kaplan-Meier survival curve of the TCGA-LUAD cohort dichotomized by the expression of COL11A1. The survival curves were compared by log-rank test. f, Spatial distribution of tumor spots, the fractions of COL11A1+ CAFs and ADH1B+ CAFs and the expression of COL11A1 and ADH1B in the slide of isolated tumor from P12. g,h, Scatter plots showing a significantly negative correlation between COL11A1+ CAF abundance and distance to tumors (spot) and a positive correlation between ADH1B+ CAF abundance and distance to tumors (spot) in the slides of isolated tumor from P11 (g) and P12 (h). The expression levels of COL11A1 or ADH1B in each spot are shown by the color gradient. The curves were fitted using a locally weighted regression (loess) model. The two-sided P values were determined by Pearson’s correlation test. i, Spatial distribution of tumor spots, fractions of COL11A1+ CAFs and ADH1B+ CAFs and the expression of COL11A1 and ADH1B in the slides of tumor stromal regions from P02 and P09. j, Scatter plot showing a significant negative correlation between the colocalization levels of COL11A1−DDR1 and the collagen formation signatures and distance to tumors (spot) in the slide from P11. The curves were fitted using linear and locally weighted regression (loess) models, respectively. The two-sided P values were determined by Pearson’s correlation test. Related to Fig.4.
Extended Data Fig. 5 Spatial transcriptomic and scRNA-seq analyses of macrophage subsets after ICB-chemotherapy.
a, Boxplots showing the fractions of Mono/Macro subsets in matched samples from 3 responders and 4 non-responders at baseline and post-treatment phase. b, Boxplots showing the fractions of Mono/Macro subsets from pre-treatment responders (PreR, n = 3), pre-treatment non-responders (PreNR, n = 4), post-treatment responders (PostR, n = 6) and post-treatment non-responders (PostNR, n = 13). c, Scattered plot showing a significant negative correlation between the fraction of SPP1+ macrophages or CXCL9+ macrophages and distance to tumors (spot) in the slide of isolated tumor from P11. The expression level of SPP1 or CXCL9 in each spot is shown by the color gradient. d, Spatial distribution of tumor spots, fractions of SPP1+ macrophages and CXCL9+ macrophages and the expression of SPP1 and CXCL9 in the slide of isolated tumor from P12. e, Scattered plot showing a significant negative correlation between the fractions of SPP1+ macrophages or CXCL9+ macrophages and the distances to tumors (spot) in the slide of isolated tumor from P12. Expression levels of SPP1 or CXCL9 in each spot are shown by the color gradient. f, Scatter plot showing a significant negative correlation between the colocalization levels of CXCL9-CXCR3 and cytotoxic signature and the distances to tumors (spot) in the slide from P11. g, Heatmap showing the averaged module scores of M1 and M2 signatures among macrophage subsets. h, Boxplots showing the fractions of CXCL9+ macrophages in matched samples from 3 responders and 4 non-responders at baseline and post-treatment. i, Box plots of average expression of CXCL9 in CXCL9+ macrophages in matched samples from 3 responders and 4 non-responders at baseline and post-treatment phase. This result indicates that CXCL9 expression in macrophages before but not post-treatment is high in responders. In panels c, e and f, the curves were fitted using locally weighted regression (loess), linear and loess models, respectively. Two-sided P values were determined by Pearson’s correlation test. In panels a, b, h and i, a one-sided t-test was used to determine the statistical significance. For the boxplots, box limits denote the first and third quartiles, with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box. Related to Fig.5.
Extended Data Fig. 6 Spatial distribution of T and B cells and identification of TLSs.
a, The spatial distribution of T cells in patient 11 (P11). The darker red color represents a higher cell-type proportion. b, The spatial distribution of B cells P11. The darker red color represents a higher cell-type proportion. c, Boxplots showing the differences in T cell subtype proportions in tumor (n = 150), tumor boundary (n = 162) and stromal (n = 3651) regions in patient P11. Statistical significance was determined by two-sided Wilcoxon rank-sum test. ns: P > = 0.05; *: P < 0.05; **: P < 0.01; ***: P < 0.001; ****: P < 0.0001. d, Heat-dot plot showing cell-type colocalizations in patient P11. Pearson correlations of the estimated cell-type proportions were calculated for each pair of cell types across all spots. The color and size of the dot represent the correlation coefficient. A positive correlation coefficient indicates colocalization of the cell-type pair, whereas negative values indicate exclusion of the two cell types from each other. The black rectangle highlights the colocalization of T and B cells. e, Pathological examination of TLSs in patient P11. Scale bar: 1 mm. All Visium slides (n = 17) were examined. f, The automated TLS identification workflow named Space Scanner through combining H&E image and ST data. g, Location of identified TLSs in patient P11. Red and gray colors represent TLSs and surrounding stroma respectively. h, Heatmap showing the averaged expression of 12 chemokines in TLSs and stroma from patient P11. i, Boxplots showing the differences in B (left panels) and T (right panels) cell subtype proportions within TLSs (n = 210) and stroma (n = 294) in patient P11. Statistical significance was determined by two-sided Wilcoxon rank-sum test. ns: P > = 0.05; *: P < 0.05; **: P < 0.01; ***: P < 0.001; ****: P < 0.0001. For the boxplots in c and i, the box limits denote the first and third quartiles, with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box.
Extended Data Fig. 7 Spatial distributions of T and B cells and identification of TLSs.
a, Spatial distributions of CD4 T cells, Treg and CD8 T cells, relative to tumor spots in patient P11. CD4 and CD8 T cell proportions were measured by the sum of the corresponding subtypes. The darker red color in the left 3 panels represents higher cell-type proportions. The right panel shows the definition of tumor spots, tumor-boundaries and stroma regions in patient P11. b, The spatial distribution of T (top) and B cell proportions (middle) and the location of identified TLSs (bottom) in samples excluding P11. c, Cell-type compositions of the identified TLSs in 14 samples. The average proportions of all TLS spots were calculated to represent the cellular compositions of each TLS. Related to Extended Data Fig.6.
Extended Data Fig. 8 Characteristics of TLSs in different stages.
a, Spatial distributions of LTB expression in all slides with TLSs. b, Boxplots showing the differences in B (left) and T (right) subtype proportions in TLSs at different stages from all samples. For each TLS, the average subtype proportions of covered spots were calculated to represent the cellular subtype proportions of each TLS. lymphoid aggregates, n = 42; Activated TLS, n = 71; Declining TLS, n = 103; Late TLS, n = 67. Statistical significance was determined by two-sided Wilcoxon rank-sum test. ns: P > = 0.05; *: P < 0.05; **: P < 0.01; ***: P < 0.001; ****: P < 0.0001. The box limits denote the first and third quartiles, with the median shown in the center and whiskers covering data within 1.5× the interquartile range from the box. c, Bar plot showing the enriched Hallmark pathways in NMPR, related to Fig. 6h. The color represents the significance. A P value denotes the enrichment P value of the pathway ID determined by Fisher’s exact test. d, PCA visualization of the hypoxia signature score in all TLSs. e, The correlation between the hypoxia signature score and the fraction of Treg (left) and CD4_TCF7 (right) in all TLSs. The blue lines are regression lines. The gray bands are 95% confidence bands for the regression line. Each dot represents an identified TLS. The two-sided P values was measured by Pearson ‘s correlation test. Related to Fig.6.
Supplementary information
Supplementary Information
Supplementary Notes 1–4.
Supplementary Tables
Supplementary Tables 1–4.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
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 57, 126–139 (2025). https://doi.org/10.1038/s41588-024-01998-y
Received:
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s41588-024-01998-y
This article is cited by
-
scXDR: drug response prediction across single-cell datasets via heterogeneous network transfer learning
Communications Biology (2026)
-
Bibliometric analysis of research trends and hotspots in immunotherapy biomarkers for non-small cell lung cancer from 2015 to 2024
Discover Oncology (2026)
-
Application of single-cell and spatial omics in deciphering cellular hallmarks of cancer drug response and resistance
Journal of Hematology & Oncology (2025)
-
Spatial-reprogramming derived GPNMB+ macrophages interact with COL6A3+ fibroblasts to enhance vascular fibrosis in glioblastoma
Genome Medicine (2025)
-
Harnessing multi-omics approaches to decipher tumor evolution and improve diagnosis and therapy in lung cancer
Biomarker Research (2025)


