Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Tumour DDR1 promotes collagen fibre alignment to instigate immune exclusion

Abstract

Immune exclusion predicts poor patient outcomes in multiple malignancies, including triple-negative breast cancer (TNBC)1. The extracellular matrix (ECM) contributes to immune exclusion2. However, strategies to reduce ECM abundance are largely ineffective or generate undesired outcomes3,4. Here we show that discoidin domain receptor 1 (DDR1), a collagen receptor with tyrosine kinase activity5, instigates immune exclusion by promoting collagen fibre alignment. Ablation of Ddr1 in tumours promotes the intratumoral penetration of T cells and obliterates tumour growth in mouse models of TNBC. Supporting this finding, in human TNBC the expression of DDR1 negatively correlates with the intratumoral abundance of anti-tumour T cells. The DDR1 extracellular domain (DDR1-ECD), but not its intracellular kinase domain, is required for immune exclusion. Membrane-untethered DDR1-ECD is sufficient to rescue the growth of Ddr1-knockout tumours in immunocompetent hosts. Mechanistically, the binding of DDR1-ECD to collagen enforces aligned collagen fibres and obstructs immune infiltration. ECD-neutralizing antibodies disrupt collagen fibre alignment, mitigate immune exclusion and inhibit tumour growth in immunocompetent hosts. Together, our findings identify a mechanism for immune exclusion and suggest an immunotherapeutic target for increasing immune accessibility through reconfiguration of the tumour ECM.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: DDR1 promotes mammary tumour growth in immunocompetent hosts.
Fig. 2: DDR1 inhibits the infiltration of anti-tumour immune cells.
Fig. 3: DDR1-dependent ECM remodelling inhibits anti-tumour immune infiltration.
Fig. 4: DDR1 as a therapeutic target for tumour immunotherapy.

Similar content being viewed by others

Data availability

DDR1 protein expression correlation scatter plots were drawn with data obtained from the CPTAC (https://cptac-data-portal.georgetown.edu/study-summary/S015) (Fig. 2e, Extended Data Fig. 3p). The correlation between DDR1 mRNA levels and patient survival was performed with data acquired from the Kaplan–Meier Plotter database (https://kmplot.com/analysis/) (Extended Data Fig. 3f, g). Correlations between the mRNA levels of DDR1 and immune markers were performed with data extracted from GSE88847 (Extended Data Fig. 3k–n) and the TCGA project (https://www.cbioportal.org/study/summary?id=brca_tcga_pan_can_atlas_2018) (Fig. 2d, Extended Data Figs. 3h–j, o, 10e). Disease-specific survival Kaplan–Meier curves were drawn with data obtained from the TCGA project (https://gdc.cancer.gov/) (Extended Data Fig 10a–d). All data generated and analysed during this study, except the RNA-seq dataset, are included in this published Article and its supplementary files. The RNA-seq dataset has been deposited to the NCBI Gene Expression Omnibus database and the accession number is GSE139239Source data are provided with this paper.

References

  1. Gruosso, T. et al. Spatially distinct tumor immune microenvironments stratify triple-negative breast cancers. J. Clin. Invest. 129, 1785–1800 (2019).

    Article  Google Scholar 

  2. Cox, T. R. The matrix in cancer. Nat. Rev. Cancer 21, 217–238 (2021).

    Article  CAS  Google Scholar 

  3. Bejarano, L., Jordao, M. J. C. & Joyce, J. A. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 11, 933–959 (2021).

    Article  CAS  Google Scholar 

  4. Chen, Y. et al. Type I collagen deletion in αSMA+ myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell 39, 548–565 (2021).

    Article  CAS  Google Scholar 

  5. Leitinger, B. Discoidin domain receptor functions in physiological and pathological conditions. Int. Rev. Cell Mol. Biol. 310, 39–87 (2014).

    Article  CAS  Google Scholar 

  6. Valiathan, R. R., Marco, M., Leitinger, B., Kleer, C. G. & Fridman, R. Discoidin domain receptor tyrosine kinases: new players in cancer progression. Cancer Metastasis Rev. 31, 295–321 (2012).

    Article  CAS  Google Scholar 

  7. Hidalgo-Carcedo, C. et al. Collective cell migration requires suppression of actomyosin at cell-cell contacts mediated by DDR1 and the cell polarity regulators Par3 and Par6. Nat. Cell Biol. 13, 49–58 (2011).

    Article  CAS  Google Scholar 

  8. Gao, H. et al. Multi-organ site metastatic reactivation mediated by non-canonical discoidin domain receptor 1 signaling. Cell 166, 47–62 (2016).

    Article  CAS  Google Scholar 

  9. Edwards, D. N. et al. Selective glutamine metabolism inhibition in tumor cells improves anti-tumor T lymphocyte activity in triple-negative breast cancer. J Clin. Invest. 131, e140100 (2021).

    Article  CAS  Google Scholar 

  10. Jiang, P. et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 24, 1550–1558 (2018).

    Article  CAS  Google Scholar 

  11. Jimenez-Sanchez, A., Cast, O. & Miller, M. L. Comprehensive benchmarking and integration of tumor microenvironment cell estimation methods. Cancer Res. 79, 6238–6246 (2019).

    Article  CAS  Google Scholar 

  12. Vogel, W. F., Aszodi, A., Alves, F. & Pawson, T. Discoidin domain receptor 1 tyrosine kinase has an essential role in mammary gland development. Mol. Cell. Biol. 21, 2906–2917 (2001).

    Article  CAS  Google Scholar 

  13. Takai, K. et al. Discoidin domain receptor 1 (DDR1) ablation promotes tissue fibrosis and hypoxia to induce aggressive basal-like breast cancers. Genes Dev. 32, 244–257 (2018).

    Article  CAS  Google Scholar 

  14. Carafoli, F. et al. Structure of the discoidin domain receptor 1 extracellular region bound to an inhibitory Fab fragment reveals features important for signaling. Structure 20, 688–697 (2012).

    Article  CAS  Google Scholar 

  15. Vogel, W. F. Ligand-induced shedding of discoidin domain receptor 1. FEBS Lett. 514, 175–180 (2002).

    Article  CAS  Google Scholar 

  16. Agarwal, G., Mihai, C. & Iscru, D. F. Interaction of discoidin domain receptor 1 with collagen type 1. J. Mol. Biol. 367, 443–455 (2007).

    Article  CAS  Google Scholar 

  17. Ruggeri, J. M. et al. Discoidin domain receptor 1 (DDR1) is necessary for tissue homeostasis in pancreatic injury and pathogenesis of pancreatic ductal adenocarcinoma. Am. J. Pathol. 190, 1735–1751 (2020).

    Article  CAS  Google Scholar 

  18. Ambrogio, C. et al. Combined inhibition of DDR1 and Notch signaling is a therapeutic strategy for KRAS-driven lung adenocarcinoma. Nat. Med. 22, 270–277 (2016).

    Article  CAS  Google Scholar 

  19. Salmon, H. et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Cell. Invest. 122, 899–910 (2021).

  20. Kaur, A. et al. Remodeling of the collagen matrix in aging skin promotes melanoma metastasis and affects immune cell motility. Cancer Discov. 9, 64–81 (2019).

  21. Ray, A. & Provenzano, P. P. Aligned forces: Origins and mechanisms of cancer dissemination guided by extracellular matrix architecture. Curr. Opin. Cell Biol. 72, 63–71 (2021).

  22. Tomko, L. A. et al. Targeted matrisome analysis identifies thrombospondin-2 and tenascin-C in aligned collagen stroma from invasive breast carcinoma. Sci. Rep. 8, 12941 (2018).

    Article  ADS  Google Scholar 

  23. Dunlap, S. M. et al. Dietary energy balance modulates epithelial-to-mesenchymal transition and tumor progression in murine claudin-low and basal-like mammary tumor models. Cancer Prev. Res. 5, 930–942 (2012).

    Article  CAS  Google Scholar 

  24. Mertins, P. et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature 534, 55–62 (2016).

    Article  CAS  Google Scholar 

  25. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  Google Scholar 

  26. Fougner, C., Bergholtz, H., Norum, J. H. & Sorlie, T. Re-definition of claudin-low as a breast cancer phenotype. Nat. Commun. 11, 1787 (2020).

    Article  ADS  CAS  Google Scholar 

  27. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  Google Scholar 

  28. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

  29. Ashburner, M. et al. Gene ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  Google Scholar 

  30. Suzuki, R. & Shimodaira, H. Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics 22, 1540–1542 (2006).

    Article  CAS  Google Scholar 

  31. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists. Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  Google Scholar 

  32. Li, T. et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 48, W509–W514 (2020).

    Article  CAS  Google Scholar 

  33. Newman, A. M. et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 37, 773–782 (2019).

    Article  CAS  Google Scholar 

  34. Erikson, A., Ortegren, J., Hompland, T., de Lange Davies, C. & Lindgren, M. Quantification of the second-order nonlinear susceptibility of collagen I using a laser scanning microscope. J. Biomed. Opt. 12, 044002 (2007).

    Article  ADS  Google Scholar 

  35. Wu, B. et al. PPARγ inhibition boosts efficacy of immune checkpoint immunotherapy against murine melanoma in a sexually dimorphic manner. Int. J. Biol. Sci. 16, 1526–1535 (2020).

    Article  CAS  Google Scholar 

  36. Meng, W. et al. Efficient generation of monoclonal antibodies from single rhesus macaque antibody secreting cells. MAbs 7, 707–718 (2015).

    Article  CAS  Google Scholar 

  37. Gui, X. et al. Disrupting LILRB4/APOE interaction by an efficacious humanized antibody reverses T-cell suppression and blocks AML development. Cancer Immunol. Res. 7, 1244–1257 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Hursting for M-Wnt cells, S. Abrams for AT-3 cells, L. Sun for HCC1937 cells, and John R Hawse and Thomas C. Spelsberg for Hs578T cells. We also thank L. Lin for technical assistance in plasmid construction, L. Audoly for discussion, G. T. Salazar for editing the manuscript and X. Zhang for technical assistance with oestrogen receptor and progesterone receptor (ER and PR) immunohistochemistry. The work was supported by grants to R.L. and T.J.C. from the National Institutes of Health (NIH) (CA206529); to R.L. from NIH (CA246707) and the Walter G. Ross Foundation; to T.J.C. from NIH (CA205965) and the Owens Foundation and the Skinner Endowment; to Y.H. from NIH (CA212674); the Congressionally Directed Medical Research Program (W81XWH-17-1-0008); to V.X.J from NIH (GM114142); and to Z.A. from the Cancer Prevention and Research Institute of Texas (RP150551 and RP190561) and the Welch Foundation (AU-0042-20030616). The Genome Sequencing Facility at the UT Health San Antonio is supported by NIH-NCI P30 CA054174 (Mays Cancer Center at UT Health San Antonio), NIH Shared Instrument grant 1S10OD021805-01 (S10 grant), and CPRIT Core Facility Award (RP160732). Georgetown University Medical Center Shared Resources are supported in part by P30 CA051008 (Lombardi Comprehensive Cancer Center Support Grant; Principal Investigator L. Weiner). The ICO-IDIBELL research was supported by the Generalitat de Catalunya (SGR 2017-449; PFI-Salut SLT017-20-000076; and CERCA program) and the Carlos III Institute of Health (ISCIII), funded by FEDER funds (‘A way to build Europe’), grants PI18/01029 and PI21/01306.

Author information

Authors and Affiliations

Authors

Contributions

R.L. managed and oversaw the overall project. R.L., X.S., Z.A., T.J.C. and N.Z. designed the experiments and wrote the manuscript. X.S., B.W., H.D., X.Z., H.-C.C., D.Z., W.X., J.L., P.M., D.B., C.I., A.M.R., M.P., D.B., B.H., C.L., K.C., P.S.L., C.A.B. and A.P. performed the experiments. X.S., B.W., M.A.P., E.B., A.G., R.E.G., D.B., C.A.B., X.Z., P.S.L., Y.Z., V.X.J., A.P., Y.H., N.Z., T.J.C., Z.A. and R.L. analysed the data.

Corresponding authors

Correspondence to Miguel Angel Pujana, Tyler J. Curiel, Zhiqiang An or Rong Li.

Ethics declarations

Competing interests

X.S., H.D., N.Z., Z.A. and R.L. are co-inventors of a pending patent application (62/949,300) filed by the University of Texas Health Science Center at Houston on the anti-DDR1 antibodies described in this manuscript. The University of Texas System and The George Washington University have licensed the patent to Parthenon Therapeutics for drug development. R.L. receives stock option and financial compensation for his role as a member on the Scientific Advisory Board of Parthenon Therapeutics.

Additional information

Peer review information Nature thanks Joan Brugge, Shannon Turley and the other, anonymous, reviewers for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Differential effects of tumour Ddr1-KO on tumour growth in vitro and in vivo.

(a) Immunoblotting of DDR1, DDR2 and loading control β-ACTIN in M-Wnt and AT-3 Ddr1-WT/KO tumour cells. Images are representatives of three independent experiments. (b–d) In vitro cell proliferation of E0771 (WT: n = 3, KO: n = 5, b), M-Wnt (WT: n = 3, KO: n = 5, c) and AT-3 (WT: n = 6, KO: n = 4, d) tumour cells, n indicate technical repeats. Out of three biological repeats. (e–f) M-Wnt (n = 4 tumours/group, e) and AT-3 (n = 5 tumours/group, f) tumour growth in immunodeficient mice. (g–h) M-Wnt (n = 7 tumours/group, g) and AT-3 (n = 7 tumours/group, h) tumour growth in immunocompetent C57BL/6 mice. (i–j) M-Wnt and AT-3 tumours were grown firstly in Rag1−/− hosts. Approximately 60 mg of tumour pieces were transplanted to C57BL/6 mice. Tumour volume of M-Wnt (WT” n = 9 tumours, KO: n = 10 tumours, i) and AT-3 (WT: n = 10 tumours, KO: n = 9 tumours, j). (k) Percentage of CD8+ in CD3+ T cells in blood, n = 5 mice/group. (l) Tumour volumes in C57BL/6 hosts with prior treatment of anti-IgG or anti-CD8 antibody (n = 5 tumours/group). (m) CD8+ TILs normalized by tumour weight in Rag1−/− mice after adoptive transfer of CD8+ T cells or medium (sham), n = 6 tumours/group. (n) Tumour volumes in Rag1−/− mice after adoptive transfer of CD8+ T cells or medium (sham). n = 6 tumours/group. Arrow indicates transfer of CD8+ T cells on day 17. (o) Tumour weight from rechallenged mice (n = 6 tumours/group). Values represent mean ± SEM. p value and n as indicated, all tests used two-way ANOVA except for CD8+ quantification, which used two-tailed Student’s t-test.

Source data

Extended Data Fig. 2 Immunophenotyping of Ddr1-WT and Ddr1-KO tumours.

(a–d) TIL number normalized by E0771 Ddr1-WT/KO tumour weight/gram. Cell number of CD44hi CD62Llo CD8+ (a) and CD44hi CD62Llo CD4+ (b) IFNγ+ CD8+ (c) and IFNγ+ CD4+ (d) T cells. WT/KO: n = 5 tumours/group. (e–p) TIL numbers normalized by tumour weight of M-Wnt (n = 4 tumours/group, e-j) and AT-3 tumours (n = 5 tumours/group, k–p). (qt) Percentages of T cells from E0771 Ddr1-WT and KO tumours (n = 5 tumours/group) positive for Ki67 (CD4+ in q and CD8+ in r), IFNγ (CD8+ in s) or GZMB (CD8+ in t). n.s. not significant. Values represent mean ± SEM. p value as indicated, two-tailed Student’s t-test.

Source data

Extended Data Fig. 3 Correlation between DDR1 and immune markers in human breast cancer.

(a) Representative images of CD8+ T cell staining at E0771 tumour margin and in the tumour core (bottom panels). (b, c) Representative images (b) and quantification (c) of CD8+ T cell IHC at M-Wnt tumour margin and core (WT: n = 8 tumours, KO: n = 4 tumours). (d, e) Representative images (d) and quantification (e) of CD8+ T cell IHC at AT-3 tumour margin and core (n = 5 tumours/group). Images in (a),(b), and (d) showing tumour margin at top panel (tumour boarder denoted by red dash lines) and tumour core at bottom panel. Box areas at higher magnification are shown in the upper right inlets. Red arrow heads indicate CD8+ cells. The y-axis in (c) and (e) refers to percent of CD8+ cells over total cells in a given field. Scale bar: 100 µm and 10 µm in inlets. Two-tailed Student’s t-test. (f–g) Correlation between DDR1 mRNA levels and overall survival of all patients with breast cancer (f) and patients with TNBC (g) in the Kaplan-Meier Plotter database (https://kmplot.com/analysis/). (h–j) Scatter plots showing the negative gene expression (Z-score) correlation between DDR1 mRNA levels and GZMB (h), IFNG (i), and PRF1 (j) in TCGA TNBC tumours (n = 162). The corresponding Spearman’s correlation coefficients and p values are shown. (k–n) Correlation of DDR1 mRNA levels and anti-tumour immune markers in 37 samples from patients with TNBC (GSE88847). (o) Scatter plot showing the negative gene expression correlation between DDR1 mRNA levels and signature for accumulation of T cells in tumours using TCGA TNBC tumour data. (p) Scatter plots showing the negative expression correlation between DDR1 protein expression and cytolytic effector pathway in CPTAC BRCA. (q) Correlation between percentages of CD8+ immune cells and DDR1+ tumour cells in a TNBC cohort (n = 12). (r) Correlation between percentages of CD8+ immune cells and DDR1+ tumour cells in a DDR1high (n = 7) and DDR1low (n = 5) TNBC samples. (s) Patient numbers of immune-excluded (n = 4) and non-immune-excluded (n = 6) in DDR1high and DDR1low group. Only the 10 patient samples with paired margin and core information were used for the immune exclusion calculation in Extended Data Fig. 3s, two-sided Chi-square test.

Source data

Extended Data Fig. 4 DDR1 dependent transcriptomic changes.

(a) Quantification of αCD31 IHC of WT and KO tumours transplanted from Rag1−/− to C57BL/6 hosts (n = 6 tumours; n.s., not significant). Data are presented as mean values +/− SEM. Two-tailed Student’s t-test (b) Comparison of non-synonymous tumour mutational burden between Ddr1-WT (n = 4 tumours) and KO tumours (n = 5 tumours, n.s., not significant). Data are presented as mean values +/− SEM. Two-tailed Student’s t-test (c) Venn diagram showing the numbers of DEGs in each Ddr1-KO-WT comparison and the identity overlaps between them. The pairwise overlap significance is indicated. The GO terms overrepresented (FDR-adjusted p < 0.05 relative to mouse genome background) in the overlapping sets are shown; the genes corresponding to each annotation are also indicated.

Source data

Extended Data Fig. 5 Mutational and biochemical analysis of DDR1-ECD in vitro and in vivo.

(a) Diagram of full-length (FL) DDR1 (top) and tumour curves of either E0771 Ddr1-WT or KO tumour cells carrying various DDR1 expression vectors: empty vector (EV), FL, deletion of the kinase domain (ΔKD), and extracellular domain (ECD) only. All p values were compared to KO + EV group. TM: transmembrane domain. WT: n = 9 tumours, KO+EV: n = 10 tumours, KO+FL: n = 10 tumours, KO+ ΔKD: n = 6 tumours, KO+ECD: n = 5 tumours. (b) Crystal structure of mouse DDR1 collagen-binding domain, generated by Jmol software (http://www.jmol.org/). Amino acid residues targeted in the mutational analysis are shown. (c) Immunoblots of Flag-tagged mouse WT DDR1-ECD and point mutants ectopically expressed in M-Wnt tumour cells, with GAPDH as the loading control. Images are representatives from three independent experiments. (d) Immunoblots of Flag-tagged mouse WT DDR1-ECD and point mutants ectopically expressed in AT-3 tumour cells, with GAPDH as the loading control. Images are representatives from three independent experiments. (e–f) Growth curves of M-Wnt (e) and AT-3 (f) Ddr1-KO tumours with ectopically expressed mouse WT DDR1-ECD or collagen-binding point mutants. The numbers in parenthesis indicate outgrowing tumours (larger than 100 mm3) versus total injected. (g, h) Immunoblots of full-length DDR1 in cells and soluble ECD in conditioned medium from various mouse (g) and triple-negative human breast cancer cell lines plus ER-positive MCF7 (h). Images are representatives from three independent experiments. (i) Coomassie staining of recombinant Fc-ECD under non-reducing and reducing conditions. (j) Rescue of Ddr1-KO E0771 tumour growth in immunocompetent hosts by recombinant Fc-ECD versus PBS vehicle (n = 6 tumours/group). (k) Diagram of the Transwell assay for CD8+ T cell migration. Primary CD8+ T cells were loaded in the upper chamber that had been pre-seeded with decellularized ECM derived from tumour cells. The lower chamber contained medium with or without CCL21. (l) CD8+ T cells in vitro migration activity was abrogated by decellularized ECM from AT-3 tumour cells in a DDR1-dependent manner. Value of migrated CD8+ T cell number without ECM and CCL12 is set at “1” (lanes 1 and 2: n = 3; lanes 3 and 4: n = 7), n refers to technical repeats. Values represent mean ± SEM. p value as indicated, two-tailed Student’s t-test for all tests except for tumour volumes, which were done by two-way ANOVA.

Source data

Extended Data Fig. 6 SHG microscopy of Ddr1-WT and Ddr1-KO tumours.

(a) E0771 Ddr1-WT/KO tumours transplanted from Rag1−/− to C57BL/6 hosts were analysed by SHG, To-pro-3 staining for all nuclei, and collagen fibre individualization. Scale bar: 50 µm. (b, c) M-Wnt (WT n = 8 tumours, KO n = 4 tumours) and AT-3 (n = 5 tumours/group) Ddr1-WT/KO tumours transplanted from Rag1−/− to C57BL/6 hosts were analysed for infiltrating CD3+ T cells normalized by total cells via IHC. (d–g) M-Wnt and AT-3 Ddr1-WT/KO tumours transplanted from Rag1−/− to C57BL/6 hosts were analysed for collagen fibre alignment (d, e) and fibre length (f, g), n = 4 tumours/group. (h–j) E0771, n = 5 tumours/group (h), M-Wnt, n = 4 tumours/group (i) and AT-3, n = 4/group (j) Ddr1-WT/KO tumours transplanted from Rag1−/− to C57BL/6 hosts were analysed for fibre numbers by the CT-Fire software. (k–m) E0771 Ddr1-WT/KO tumours (WT n = 10 tumours, KO n = 8 tumours) from immunodeficient Rag1−/− hosts were analysed for collagen fibre alignment (k), fibre length (l) and fibre numbers (m) by the CT-Fire software. (n) Growth curves of E0771 Ddr1-KO tumours in immunocompetent hosts that were intratumorally injected with recombinant WT and mutant Fc-ECD (WT: n = 10 tumours, W54A: n = 9 tumours). (o) Representative images of E0771 Ddr1-KO tumours treated with recombinant WT or mutant Fc-ECD in C57BL/6 hosts as analysed by SHG, To-pro-3 staining, and collagen fibre individualization. Scale bar: 50 µm. (p) Quantification of collagen fibre alignment in WT and mutant Fc-ECD treated tumours (n = 5 tumours/group). (q) Enumeration of infiltrating CD3+ T cells normalized by total cells via IHC (WT: n = 4 tumours, KO: n = 3 tumours). Values represent mean ± SEM. p value as indicated, two-tailed Student’s t-test for all tests except for tumour volumes, which were done by two-way ANOVA.

Source data

Extended Data Fig. 7 Screening for huDDR1-neutralizing antibodies.

(a) Immunoblots of ectopic human (hu) DDR1 and endogenous mouse DDR1 in cell lysates and medium of E0771-derived cells. (b) Tumour growth curve of E0771-derived Ddr1-WT, KO+EV, KO+huDDR1 cells (n = 7 tumours/group).(c) Transwell migration assay for purified CD8+ T cells in the presence of conditioned medium from E0771 cells containing endogenous WT DDR1, Ddr1-KO, or Ddr1-KO and ectopic expression of huDDR1 (n = 3 technical repeats). Value of migrated CD8+ T cell number with parental E0771-conditioned medium is set at “1”. (d) Quantification of CD8+ T cell migration in the presence of DDR1-neutralizing antibodies, using conditioned medium from E0771 Ddr1-KO or KO+huDDR1 cells (IgG: n = 4, #3,#9,#14,#33: n = 2, technical repeats). Control: isotype IgG; anti-DDR1 antibody: #3, #9, #14, and #33. Value of migrated CD8+ T cell number in the far-left column is set at “1”. (e) Tumour curves treated with control IgG, #3, #9, #14, and #33 (n = 8 tumours/group). Antibody administration started when tumour volume reached approximately 100 mm3. All p values were compared to the control IgG group and p value as indicated. (f) Tumours host survival curves of E0771 Ddr1-KO tumour cells with ectopically expressed human (hu) DDR1 in C57BL/6 hosts treated intratumorally with isotype IgG (Ctrl, n = 17, tumours) or anti-DDR1 antibody #9 (n = 18, tumours). (g) Host body weight treated with control IgG, #3, #9, #14, and #33 (Ctrl n = 4 mice, #3, #9, #14, and #33 n = 4 mice/group). Antibody administration started when tumour volume reached approximately 100 mm3. Data are presented as mean values +/− SEM. (h, i) E0771 KO+huDDR1 tumours in C57BL/6 (n = 8 tumours/group, h) and Rag1−/− hosts (n = 6 tumours/group, i) treated with either isotype IgG or anti-DDR1 #33 antibody. (j, k) Tumour volume (j) and survival curve (k) of M-Wnt KO+huDDR1 tumours in C57BL/6 mice treated with isotype IgG and anti-DDR1 antibody #9 (n = 10, tumours/group). (l, m) Tumour growth (l) and survival percentage (m) of AT-3 KO+huDDR1 tumours in C57BL/6 mice treated with isotype IgG and anti-DDR1 antibody #3 (n = 10, tumours/group). Values represent mean ± SEM. p value as indicated. Tumour volumes were examined by two-way ANOVA; survival analysis was examined by log-rank (Mantel–Cox) test, and migration assay were examined by two-tailed Student’s t-test.

Source data

Extended Data Fig. 8 DDR1 antibody treatment inhibits spontaneous mammary tumour growth.

(a-b) Binding affinity of all four anti-ECD antibody clones for human (a) and mouse (b) DDR1. (c–d) Spontaneous MMTV-PyMT body weight of C57BL/6 hosts treated with control or #9 in the pre-tumour (from 11 weeks old, control: n = 7 mice, #9: n = 8 mice, c) and post-tumour groups (control n = 7 mice, #9 n = 8 mice, d). Data are presented as mean values +/− SEM. (e) Tumour incidence (percentage of tumour-bearing mammary glands per mouse, in MMTV-PyMT spontaneous mammary tumour model of C57BL/6 genetic background, treated in a “post-tumour” scheme with Ctrl (n = 7 mice) or anti-DDR1 #9 antibody (n = 8 mice). Data are presented as mean values + SEM. (f, g) Spontaneous MMTV-PyMT tumour growth in C57BL/6 hosts (accumulative tumour volume per mouse, f) and incidence percentage (per mouse, g) with pre-tumour treatment (control: n = 7 mice, #9: n = 8 mice). Data are presented as mean values +/− SEM. (h, i) Spontaneous MMTV-PyMT tumour growth (accumulative tumour volume per mouse) in FVB hosts treated with control or #9 before tumour growth (from 5 to 7 weeks old, h) and post-tumour groups (i). control: n = 7 mice, #9: n = 8 mice, n.s. not significant. Two-way ANOVA were used for all tests.

Source data

Extended Data Fig. 9 DDR1 antibody boosts the infiltration of anti-tumour immune cells.

(a–d) Indicated TIL numbers normalized by tumour weight in E0771 KO+huDDR1 tumours from C57BL/6 mice treated with control and anti-DDR1 antibody #9 (n = 4 tumours/group). (e–h) Percentage of Ki67-positive cells in CD4+, CD8+ T cells and percentage of IFNγ- or GZMB-positive cells in CD8+ T cells from the same antibody-treated mice as in (a–d) (n = 4 tumours/group). n.s. not significant. (i) Representative images of transplanted mammary tumours treated with Ctrl or anti-DDR1 #9 antibody, analysed by SHG, To-pro-3 staining, and collagen fibre individualization. Scale bar: 50 µm. (j) Quantification of CD8+ T cells in tumour margin and core in control and anti-DDR1 antibody-treated E0771 KO+huDDR1 tumours (Ctrl: n = 8 tumours, #9: n = 9 tumours). (k, l) TILs from spontaneous mammary tumours (C57B/6) treated with Ctrl or anti-DDR1 #9 antibody under the pre-tumour (k) and post-tumour (l) conditions. n = 6 tumours/group. (m, n) Representative IHC images of CD3+ and CD8+ T cells in tumour margin and core in control and anti-DDR1 antibody-treated E0771 KO+huDDR1 tumours. Tumour boarder denoted by red dash lines. Box areas at higher magnification are shown in the inlets. Red arrow heads indicate CD8+ cells. Scale bar: 100 µm and 10 µm in inlet. (o) Representative images of tumours from the post-tumour treatment group, analysed by SHG, To-pro-3 staining, and collagen fibre individualization. Scale bar: 50 µm. (p) Quantification of tumour fibre alignment in pre- and post-tumour treatment in C57BL/6 hosts (n = 5 tumours/group). Values represent mean ± SEM. p value as indicated, two-tailed Student’s t-test.

Source data

Extended Data Fig. 10 DDR1-related clinical correlation in cancers.

(a–d) Kaplan–Meier curves showing disease specific survival (DSS) rates for TCGA patients with breast cancer divided by major tumour subtypes: basal-like (a), HER2 positive (b), luminal A (c), and luminal B (d). Each subtype is further divided in four patient groups according to the tumour expression levels of the DDR1 gene and collagen-alignment signature. The gene/signature classification in high and low expression was based on their corresponding average expression values. The log-rank test p value and the number of individuals at risk at different follow-up times are shown in each tumour subtype analysis. (e) Correlation between human DDR1 and GZMB mRNA expression in various cancer types.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1–14 and their accompanying legends.

Reporting Summary

Supplementary Table 1

Most variable gene expression and associated GO terms.

Supplementary Table 2

DEGs between Ddr1-KO and Ddr1-WT tumours.

Supplementary Table 3

Cell types and differences between Ddr1-KO and Ddr1-WT tumours in immunocompetent host.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, X., Wu, B., Chiang, HC. et al. Tumour DDR1 promotes collagen fibre alignment to instigate immune exclusion. Nature 599, 673–678 (2021). https://doi.org/10.1038/s41586-021-04057-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-04057-2

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer