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.

  • Resource
  • Published:

Tumor-induced stromal reprogramming drives lymph node transformation

Abstract

Lymph node (LN) stromal cells, particularly fibroblastic reticular cells (FRCs), provide critical structural support and regulate immunity, tolerance and the transport properties of LNs. For many tumors, metastasis to the LNs is predictive of poor prognosis. However, the stromal contribution to the evolving microenvironment of tumor-draining LNs (TDLNs) remains poorly understood. Here we found that FRCs specifically of TDLNs proliferated in response to tumor-derived cues and that the network they formed was remodeled. Comparative transcriptional analysis of FRCs from non-draining LNs and TDLNs demonstrated reprogramming of key pathways, including matrix remodeling, chemokine and/or cytokine signaling, and immunological functions such as the recruitment, migration and activation of leukocytes. In particular, downregulation of the expression of FRC-derived chemokine CCL21 and cytokine IL-7 were accompanied by altered composition and aberrant localization of immune-cell populations. Our data indicate that following exposure to tumor-derived factors, the stroma of TDLNs adapts on multiple levels to exhibit features typically associated with immunosuppression.

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

Figure 1: LN expansion and FRC remodeling.
Figure 2: Statistical analysis of microarray results.
Figure 3: Identification of specific genes and pathways deregulated in TDLN FRCs.
Figure 4: Perturbation in chemokine and cytokine signaling alters the composition and localization of immune cells.
Figure 5: FRCs in TDLNs are activated.
Figure 6: Modified transporter repertoires within TDLN FRCs translate into altered solute transport throughout the conduit system.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Bajénoff, M. et al. Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes. Immunity 25, 989–1001 (2006).

    PubMed  PubMed Central  Google Scholar 

  2. Link, A. et al. Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells. Nat. Immunol. 8, 1255–1265 (2007).

    CAS  PubMed  Google Scholar 

  3. Schumann, K. et al. Immobilized chemokine fields and soluble chemokine gradients cooperatively shape migration patterns of dendritic cells. Immunity 32, 703–713 (2010).

    CAS  PubMed  Google Scholar 

  4. Roozendaal, R., Mebius, R.E. & Kraal, G. The conduit system of the lymph node. Int. Immunol. 20, 1483–1487 (2008).

    CAS  PubMed  Google Scholar 

  5. Gretz, J.E., Norbury, C.C., Anderson, A.O., Proudfoot, A.E. & Shaw, S. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J. Exp. Med. 192, 1425–1440 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Roozendaal, R. et al. Conduits mediate transport of low-molecular-weight antigen to lymph node follicles. Immunity 30, 264–276 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Sixt, M. et al. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 22, 19–29 (2005).

    CAS  PubMed  Google Scholar 

  8. Luther, S.A., Tang, H.L., Hyman, P.L., Farr, A.G. & Cyster, J.G. Coexpression of the chemokines ELC and SLC by T zone stromal cells and deletion of the ELC gene in the plt/plt mouse. Proc. Natl. Acad. Sci. USA 97, 12694–12699 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Förster, R., Davalos-Misslitz, A.C. & Rot, A. CCR7 and its ligands: balancing immunity and tolerance. Nat. Rev. Immunol. 8, 362–371 (2008).

    PubMed  Google Scholar 

  10. Katakai, T. et al. A novel reticular stromal structure in lymph node cortex: an immuno-platform for interactions among dendritic cells, T cells and B cells. Int. Immunol. 16, 1133–1142 (2004).

    CAS  PubMed  Google Scholar 

  11. Katakai, T., Hara, T., Sugai, M., Gonda, H. & Shimizu, A. Lymph node fibroblastic reticular cells construct the stromal reticulum via contact with lymphocytes. J. Exp. Med. 200, 783–795 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Gunn, M.D. et al. A chemokine expressed in lymphoid high endothelial venules promotes the adhesion and chemotaxis of naive T lymphocytes. Proc. Natl. Acad. Sci. USA 95, 258–263 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Fletcher, A.L. et al. Lymph node fibroblastic reticular cells directly present peripheral tissue antigen under steady-state and inflammatory conditions. J. Exp. Med. 207, 689–697 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Lee, J.W. et al. Peripheral antigen display by lymph node stroma promotes T cell tolerance to intestinal self. Nat. Immunol. 8, 181–190 (2007).

    CAS  PubMed  Google Scholar 

  15. Nichols, L.A. et al. Deletional self-tolerance to a melanocyte/melanoma antigen derived from tyrosinase is mediated by a radio-resistant cell in peripheral and mesenteric lymph nodes. J. Immunol. 179, 993–1003 (2007).

    CAS  PubMed  Google Scholar 

  16. Mueller, S.N. et al. Viral targeting of fibroblastic reticular cells contributes to immunosuppression and persistence during chronic infection. Proc. Natl. Acad. Sci. USA 104, 15430–15435 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. de Boer, M., van Dijck, J.A., Bult, P., Borm, G.F. & Tjan-Heijnen, V.C. Breast cancer prognosis and occult lymph node metastases, isolated tumor cells, and micrometastases. J. Natl. Cancer Inst. 102, 410–425 (2010).

    CAS  PubMed  Google Scholar 

  18. Morton, D.L. et al. MSLT Group. Sentinel-node biopsy or nodal observation in melanoma. N. Engl. J. Med. 355, 1307–1317 (2006).

    CAS  PubMed  Google Scholar 

  19. Karaman, S. & Detmar, M. Mechanisms of lymphatic metastasis. J. Clin. Invest. 124, 922–928 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Malhotra, D. et al. Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks. Nat. Immunol. 13, 499–510 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Cox, T.R. & Erler, J.T. Molecular pathways: connecting fibrosis and solid tumor metastasis. Clin. Cancer Res. 20, 3637–3643 (2014).

    CAS  PubMed  Google Scholar 

  22. Kalluri, R. & Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 6, 392–401 (2006).

    CAS  PubMed  Google Scholar 

  23. Cremasco, V. et al. B cell homeostasis and follicle confines are governed by fibroblastic reticular cells. Nat. Immunol. 15, 973–981 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Fletcher, A.L., Acton, S.E. & Knoblich, K. Lymph node fibroblastic reticular cells in health and disease. Nat. Rev. Immunol. 15, 350–361 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Förster, R., Braun, A. & Worbs, T. Lymph node homing of T cells and dendritic cells via afferent lymphatics. Trends Immunol. 33, 271–280 (2012).

    PubMed  Google Scholar 

  26. Gunn, M.D. et al. Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J. Exp. Med. 189, 451–460 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Mueller, S.N. et al. Regulation of homeostatic chemokine expression and cell trafficking during immune responses. Science 317, 670–674 (2007).

    CAS  PubMed  Google Scholar 

  28. Yang, C.Y. et al. Trapping of naive lymphocytes triggers rapid growth and remodeling of the fibroblast network in reactive murine lymph nodes. Proc. Natl. Acad. Sci. USA 111, E109–E118 (2014).

    CAS  PubMed  Google Scholar 

  29. Harrell, M.I., Iritani, B.M. & Ruddell, A. Tumor-induced sentinel lymph node lymphangiogenesis and increased lymph flow precede melanoma metastasis. Am. J. Pathol. 170, 774–786 (2007).

    PubMed  PubMed Central  Google Scholar 

  30. Hirakawa, S. et al. VEGF-C-induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites. Blood 109, 1010–1017 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Hirakawa, S. et al. VEGF-A induces tumor and sentinel lymph node lymphangiogenesis and promotes lymphatic metastasis. J. Exp. Med. 201, 1089–1099 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Rohner, N.A. et al. Lymph node biophysical remodeling is associated with melanoma lymphatic drainage. FASEB J. 29, 4512–4522 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Carrière, V. et al. Cancer cells regulate lymphocyte recruitment and leukocyte-endothelium interactions in the tumor-draining lymph node. Cancer Res. 65, 11639–11648 (2005).

    PubMed  Google Scholar 

  34. Soudja, S.M. et al. Disrupted lymph node and splenic stroma in mice with induced inflammatory melanomas is associated with impaired recruitment of T and dendritic cells. PLoS One 6, e22639 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. MartIn-Fontecha, A. et al. Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming. J. Exp. Med. 198, 615–621 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. St. John, A.L. & Abraham, S.N. Salmonella disrupts lymph node architecture by TLR4-mediated suppression of homeostatic chemokines. Nat. Med. 15, 1259–1265 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Acton, S.E. et al. Podoplanin-rich stromal networks induce dendritic cell motility via activation of the C-type lectin receptor CLEC-2. Immunity 37, 276–289 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Astarita, J.L. et al. The CLEC-2-podoplanin axis controls the contractility of fibroblastic reticular cells and lymph node microarchitecture. Nat. Immunol. 16, 75–84 (2015).

    CAS  PubMed  Google Scholar 

  39. Augsten, M. et al. Cancer-associated fibroblasts expressing CXCL14 rely upon NOS1-derived nitric oxide signaling for their tumor-supporting properties. Cancer Res. 74, 2999–3010 (2014).

    CAS  PubMed  Google Scholar 

  40. Erez, N., Truitt, M., Olson, P., Arron, S.T. & Hanahan, D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-κνB-dependent manner. Cancer Cell 17, 135–147 (2010).

    CAS  PubMed  Google Scholar 

  41. Harper, J. & Sainson, R.C. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin. Cancer Biol. 25, 69–77 (2014).

    CAS  PubMed  Google Scholar 

  42. Kraman, M. et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 330, 827–830 (2010).

    CAS  PubMed  Google Scholar 

  43. Shields, J.D., Kourtis, I.C., Tomei, A.A., Roberts, J.M. & Swartz, M.A. Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science 328, 749–752 (2010).

    CAS  PubMed  Google Scholar 

  44. Baxter, L.T. & Jain, R.K. Transport of fluid and macromolecules in tumors. I. Role of interstitial pressure and convection. Microvasc. Res. 37, 77–104 (1989).

    CAS  PubMed  Google Scholar 

  45. Dafni, H., Landsman, L., Schechter, B., Kohen, F. & Neeman, M. MRI and fluorescence microscopy of the acute vascular response to VEGF165: vasodilation, hyper-permeability and lymphatic uptake, followed by rapid inactivation of the growth factor. NMR Biomed. 15, 120–131 (2002).

    CAS  PubMed  Google Scholar 

  46. Goldman, J. et al. Cooperative and redundant roles of VEGFR-2 and VEGFR-3 signaling in adult lymphangiogenesis. FASEB J. 21, 1003–1012 (2007).

    CAS  PubMed  Google Scholar 

  47. Hompland, T., Ellingsen, C., Øvrebø, K.M. & Rofstad, E.K. Interstitial fluid pressure and associated lymph node metastasis revealed in tumors by dynamic contrast-enhanced MRI. Cancer Res. 72, 4899–4908 (2012).

    CAS  PubMed  Google Scholar 

  48. Dankort, D. et al. Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 41, 544–552 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Gentleman, R.C. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80 (2004).

    PubMed  PubMed Central  Google Scholar 

  50. Du, P., Kibbe, W.A. & Lin, S.M. lumi: a pipeline for processing Illumina microarray. Bioinformatics 24, 1547–1548 (2008).

    CAS  PubMed  Google Scholar 

  51. Saeed, A.I. et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34, 374–378 (2003).

    CAS  PubMed  Google Scholar 

  52. Mootha, V.K. et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    CAS  PubMed  Google Scholar 

  53. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Hirosue, S. et al. Steady-state antigen scavenging, cross-presentation, and CD8+ T cell priming: a new role for lymphatic endothelial cells. J. Immunol. 192, 5002–5011 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Warde-Farley, D. et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 38, W214–W220 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Montojo, J., Zuberi, K., Rodriguez, H., Bader, G.D. & Morris, Q. GeneMANIA: Fast gene network construction and function prediction for Cytoscape. F1000 Res. 3, 153 (2014).

    Google Scholar 

Download references

Acknowledgements

We thank members of the Shields Group for comments and discussions; members of the Ares Facility QU, E23 and T29 staff for animal husbandry and technical assistance; R. Butler for support with image analysis and algorithm development; Cambridge Genomic Services for microarray services and post-analysis advice; and the CIMR flow cytometry core facility for advice and support in flow cytometry and cell-sorting applications. Supported by Medical Research Council core funding (J.S.) and the Royal Society (UF130039 to B.A.H.).

Author information

Authors and Affiliations

Authors

Contributions

A.R. planned and performed most experiments and associated analyses; D.S. performed in silico analysis; L.H. performed in vitro experiments; B.A.H. contributed to in silico analysis and data interpretation; J.S. conceived of the project, planned and performed experiments and contributed to data interpretation; A.R., D.S. and J.S. wrote the paper; and all authors contributed to editing of manuscript and critical review.

Corresponding authors

Correspondence to Benjamin A Hall or Jacqueline Shields.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 LN expansion in response to tumor-cell injection.

(a) Confocal images of B16.F10 brachial NDLNs (left), day 4 TDLNs (middle) and day 11 TDLNs (right) stained for ER-TR7 (green), CD31 (red) and LYVE1 (blue). (b) qRT-PCR quantification Tyrp1 and Dct in LN suspensions (106 cells) spiked with indicated numbers of B16.F10; or whole NDLNs, day 4 TDLNs and day 11 TDLNs. (c) Flow cytometry measurement of total LN cells, BECs, LECs, and FRCs in axillary LN (B16.F10). (d) Confocal image of NDLN (left) and TDLN (right) from TyrCreERBrafCAPtenlox stained for CD31 (green), podoplanin (red) and LYVE-1 (blue). (e) Flow cytometry quantification of EdU-labeled stromal cells. Scale bars (a) 150 µm, (d) 200 µm. Data points indicate the mean ± s.e.m. *P <0.05 (two-tailed unpaired t-test (e)). Data represent one experiment performed with n=4 for LNs (b); two independent experiments n=4 NDLNs, n=6 TDLNs in C57BL/6 female mice (c), n=6 day 2, n=6 day 4 and n=10 day 11 for both NDLNs and TDLNs in C57BL/6 female mice (e).

Supplementary Figure 2 Stromal expansion in TDLNs.

(a) Flow cytometry measurement of FRC number in TDLN. Red line denotes baseline FRC percentage/LN. (b) Confocal images of FRC networks in NDLNs (top) and day 11 TDLNs (bottom) LNs stained for podoplanin (red) and collagen I (blue). (c) Confocal Airyscans of conduit side and end views from NDLNs (top) and day 11 TDLNs (bottom) stained for podoplanin (green) ER-TR7 (blue) and CCL21 (red). (d) Confocal Airyscans of conduit side and end views from NDLNs (top) and day 11 TDLNs (bottom) stained for podoplanin (green) ER-TR7 (red) and collagen I (blue). (e) Quantification of network branch length. (f-h) Scatterplots showing the log2 fold expression change for all genes in the array (x axis), ranked according to increasing expression value (y axis); NDLNs vs. day 4 TDLNs (f), NDLNs vs. day 11 TDLNs (g), day 4 TDLNs vs. day 11 TDLNs (h). (i) Primary eigenvectors for Principal Component Analysis. The vast majority of change in the data (93.4%) are contained within the first 2 eigenvalues. Scale bars (b) 50 µm, (c) 0.487 µm (NDLN end), 0.291 µm (NDLN side), 0.588 µm (TDLN end), 0.873 µm (TDLN side), (d) 0.437 µm, 0.441 µm, 0.256 µm, 0.328 µm (NDLN end) and 0.454 µm (NDLN side); 0.363 µm, 0.481 µm, 0.47 µm, 0.363 µm (TDLN end) and 0.55 µm (TDLN side). Each symbol represents an individual FOV (e). Small horizontal lines indicate mean ± s.e.m. *P <0.05 (two-tailed unpaired t-test (a)). Data represent two independent experiments n=4 NDLNs, n= 6 TDLNs (mean ± s.e.m. (a)); or three independent experiments in female C57BL/6 mice, n=6 NDLNs and n=4 TDLNs.

Supplementary Figure 3 Overall significantly deregulated probes and pathways.

(a) Heatmap displaying all gene probes with a cutoff of change in expression of 1.5-fold for day and day 11 TDLNs. (b) Heatmap displaying normalized enrichment scores (NES) above 1.5 and with a P<0.05 for deregulated Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways using GSEA. (c) Heatmap of z-scores (indicating activation or inhibition) of canonical pathways Ingenuity Pathway Analysis (IPA) above 0.5 and P<0.05 (left) and top canonical pathways (IPA) ordered according to their P values (right). (d) Heatmap displaying z-scores (indicating activation or inhibition) of disease and biofunctions (IPA) above 2 and P<0.05 (left), and biofunctions (IPA) ordered according to their P values (right). Grey boxes represent pathways/functions with no significant deregulation.

Supplementary Figure 4 Immune-cell composition and localization in TDLNs.

(a) T cell area (left) and B cell follicle size (right) as percentage/LN measured based on CD3εe+ or CD45R+ staining. (b) Flow cytometry of CD8α+ T cells (CD45+CD3ε+CD8α +) in NDLNs and day 11 TDLNs. (c) Confocal image of NDLN (top) and day 11 TDLN (bottom) stained for FoxP3 (green) CD3ε (red) and collagen I (blue). (d) Confocal image of NDLN (left), day 4 TDLN (middle) and day 11 TDLN (right) stained for CD3ε (green), CD45R (red) and collagen I (blue). (e) Confocal image of NDLN (top) and day 11 TDLN (bottom) of paracortical area (left) or B cell follicles (right) stained for EdU (green), CD45R (red), LYVE1 (blue), and CD3ε (magenta). (f) Flow cytometry quantification of CD45+EdU+ cells in NDLNs and day 11 TDLNs. Scale bars (c) 150 µm (d and e) 50 µm. Each symbol represents an individual LN (a,b,f). Small horizontal lines indicate mean ± s.e.m. *P <0.05, **P <0.01 (two-tailed unpaired t-test (a,f)). Data represent 3 independent experiments, n=6 NDLNs and n=5 TDLNs (a), n=6 NDLNs and n=6 TDLNs (b), n=8 NDLNs and n=12 TDLNs.

Supplementary Figure 5 Macrophage and dendritic cell populations in TDLNs and verification.

(a) Confocal images of NDLNs (left) and day 11 TDLNs (right) stained for Macrophages (red) and collagen I (blue). (b) Flow cytometry quantification of CD11b+ cells (CD45+CD3ε-CD45R-CD11c-CD11b+) in NDLNs and TDLNs. (c) Flow cytometry quantification of CD11c+ cells (CD45+CD3ε-CD45R-CD11c+) in NDLNs and TDLNs. (d) qRT-PCR validation of Cd248 in an independent FRC sample set from B16.F10 NDLNs, day 4 TDLNs and day 11 TDLNs. (e) Collagen gel contraction (related to Fig. 5g). (f) qRT-PCR validation of Aqp1 in an independent FRC sample set from B16.F10 NDLNs, day 4 TDLNs and day 11 TDLNs. Scales bars 150 µm (a). Each symbol represents an individual LN (b-d,f). Small horizontal lines indicate mean ± s.e.m. *P <0.05, **P <0.01, ***P <0.001 and ****P <0.0001 (two-tailed unpaired t-test (b,c), one-way ANOVA with Tukey post hoc (d,f)). Data represent 2 independent experiments n=6 NDLNs and n=8 TDLNs (day 11) or one experiment n=4 day 4, 7 and 14 TDLNs (b,c); one experiment in technical duplicates, n=3 per condition. All in C57BL/6 female mice.

Supplementary Figure 6 Gene-interaction networks for significantly deregulated gene probes from day-11 TDLNs.

The MANIA algorithm includes 3 edge types between nodes: Predicted Edges, which implies functional relationships due to orthology with other organisms (gold), Co-localization Edges, where linked gene products are expressed in the same cellular location (blue), and Co-expression edges, where the expression levels of gene products are similar across conditions in a previously published gene expression study (pink). Node color also indicates whether a gene is predicted to be involved in the network (grey), or is one of the input genes (blue). Genes are linked through calculated edges, and networks of linked genes generated. The four most relevant probe groups are shown; all are involved in cell structure or shape. Models predict a large degree of relatedness and significance between Extracellular Matrix Proteins, Microtubule Cytoskeletal Elements, and Microtubule Regulation. Less dense networks (Structural Proteins) indicate less specific functional annotation.

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Riedel, A., Shorthouse, D., Haas, L. et al. Tumor-induced stromal reprogramming drives lymph node transformation. Nat Immunol 17, 1118–1127 (2016). https://doi.org/10.1038/ni.3492

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3492

This article is cited by

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