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.

  • Review Article
  • Published:

Metastatic niche functions and therapeutic opportunities

Abstract

Metastasis is an inefficient process, especially during colonization at a distant organ. This bottleneck underlies the importance of the metastatic niche for seeding and outgrowth of metastases. Here, we classify the common functions of different metastatic niches: anchorage, survival support, protection from external insults, licensing proliferation and outgrowth. We highlight the emerging role of the metastatic niche in maintaining cancer stemness and promoting immune evasion, and discuss therapeutic opportunities against the metastatic niche.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: The four pillars of metastatic niche functions.
Fig. 2: Niche regulation of stemness, EMT/MET and cell plasticity in distant sites.
Fig. 3: Therapeutic targeting of the metastatic niche functions.

Similar content being viewed by others

References

  1. Schofield, R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 4, 7–25 (1978).

    PubMed  CAS  Google Scholar 

  2. Scadden, D. T. Nice neighborhood: emerging concepts of the stem cell niche. Cell 157, 41–50 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Blanpain, C. & Fuchs, E. Stem cell plasticity. Plasticity of epithelial stem cells in tissue regeneration. Science 344, 1242281 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Barcellos-Hoff, M. H., Lyden, D. & Wang, T. C. The evolution of the cancer niche during multistage carcinogenesis. Nat. Rev. Cancer 13, 511–518 (2013).

    Article  PubMed  CAS  Google Scholar 

  5. Plaks, V., Kong, N. & Werb, Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 16, 225–238 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Celia-Terrassa, T. & Kang, Y. Distinctive properties of metastasis-initiating cells. Genes Dev. 30, 892–908 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Oskarsson, T., Batlle, E. & Massague, J. Metastatic stem cells: sources, niches, and vital pathways. Cell Stem Cell 14, 306–321 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Psaila, B. & Lyden, D. The metastatic niche: adapting the foreign soil. Nat. Rev. Cancer 9, 285–293 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Sleeman, J. P. The metastatic niche and stromal progression. Cancer Metast. Rev. 31, 429–440 (2012).

    Article  CAS  Google Scholar 

  10. Peinado, H. et al. Pre-metastatic niches: organ-specific homes for metastases. Nat. Rev. Cancer 17, 302–317 (2017).

    Article  PubMed  CAS  Google Scholar 

  11. Hoshino, A. et al. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Kaplan, R. N. et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438, 820–827 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Shiozawa, Y. et al. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298–1312 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Vanharanta, S. & Massague, J. Origins of metastatic traits. Cancer Cell 24, 410–421 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Headley, M. B. et al. Visualization of immediate immune responses to pioneer metastatic cells in the lung. Nature 531, 513–517 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Valiente, M. et al. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell 156, 1002–1016 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Alexander, S., Weigelin, B., Winkler, F. & Friedl, P. Preclinical intravital microscopy of the tumour-stroma interface: invasion, metastasis, and therapy response. Curr. Opin. Cell Biol. 25, 659–671 (2013).

    Article  PubMed  CAS  Google Scholar 

  18. Mayorca-Guiliani, A. E. et al. ISDoT: in situ decellularization of tissues for high-resolution imaging and proteomic analysis of native extracellular matrix. Nat. Med. 23, 890–898 (2017).

    Article  PubMed  CAS  Google Scholar 

  19. Kienast, Y. et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 16, 116–122 (2010).

    Article  PubMed  CAS  Google Scholar 

  20. Olmeda, D. et al. Whole-body imaging of lymphovascular niches identifies pre-metastatic roles of midkine. Nature 546, 676–680 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Malanchi, I. et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481, 85–89 (2012).

    Article  CAS  Google Scholar 

  22. Qian, B. et al. A distinct macrophage population mediates metastatic breast cancer cell extravasation, establishment and growth. PLoS ONE 4, e6562 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Qian, B. Z. et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475, 222–225 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Costa-Silva, B. et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17, 816–826 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Priego, N. et al. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat. Med. https://doi.org/10.1038/s41591-018-0044-4 (2018).

  26. Zhang, L. et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 527, 100–104 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. van der Weyden, L. et al. Genome-wide in vivo screen identifies novel host regulators of metastatic colonization. Nature 541, 233–236 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Price, T. T. et al. Dormant breast cancer micrometastases reside in specific bone marrow niches that regulate their transit to and from bone. Sci. Transl Med. 8, 340ra373 (2016).

    Article  Google Scholar 

  29. Ghajar, C. M. et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 15, 807–817 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Correa, D., Somoza, R. A., Lin, P., Schiemann, W. P. & Caplan, A. I. Mesenchymal stem cells regulate melanoma cancer cells extravasation to bone and liver at their perivascular niche. Int. J. Cancer 138, 417–427 (2016).

    Article  PubMed  CAS  Google Scholar 

  31. Grange, C. et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 71, 5346–5356 (2011).

    Article  PubMed  CAS  Google Scholar 

  32. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Hiratsuka, S. et al. Endothelial focal adhesion kinase mediates cancer cell homing to discrete regions of the lungs via E-selectin up-regulation. Proc. Natl Acad. Sci. USA 108, 3725–3730 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Hiratsuka, S. et al. The S100A8-serum amyloid A3-TLR4 paracrine cascade establishes a pre-metastatic phase. Nat. Cell Biol. 10, 1349–1355 (2008).

    Article  PubMed  CAS  Google Scholar 

  35. Muller, A. et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 410, 50–56 (2001).

    Article  PubMed  CAS  Google Scholar 

  36. Seguin, L., Desgrosellier, J. S., Weis, S. M. & Cheresh, D. A. Integrins and cancer: regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol. 25, 234–240 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Yu, Q., Toole, B. P. & Stamenkovic, I. Induction of apoptosis of metastatic mammary carcinoma cells in vivo by disruption of tumor cell surface CD44 function. J. Exp. Med. 186, 1985–1996 (1997).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Murgai, M. et al. KLF4-dependent perivascular cell plasticity mediates pre-metastatic niche formation and metastasis. Nat. Med. 23, 1176–1190 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Nielsen, S. R. et al. Macrophage-secreted granulin supports pancreatic cancer metastasis by inducing liver fibrosis. Nat. Cell Biol. 18, 549–560 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Erler, J. T. et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell 15, 35–44 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Hiratsuka, S. et al. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2, 289–300 (2002).

    Article  PubMed  CAS  Google Scholar 

  42. Descot, A. & Oskarsson, T. The molecular composition of the metastatic niche. Exp. Cell Res. 319, 1679–1686 (2013).

    Article  PubMed  CAS  Google Scholar 

  43. Gilkes, D. M. et al. Procollagen lysyl hydroxylase 2 is essential for hypoxia-induced breast cancer metastasis. Mol. Cancer Res. 11, 456–466 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Delloye-Bourgeois, C. et al. Microenvironment-driven shift of cohesion/detachment balance within tumors induces a switch toward metastasis in neuroblastoma. Cancer Cell 32, 427–443 (2017).

    Article  PubMed  CAS  Google Scholar 

  45. Chiarugi, P. & Giannoni, E. Anoikis: a necessary death program for anchorage-dependent cells. Biochem. Pharmacol. 76, 1352–1364 (2008).

    Article  PubMed  CAS  Google Scholar 

  46. Chen, Q., Zhang, X. H. & Massague, J. Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell 20, 538–549 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Zhang, X. H. et al. Latent bone metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell 16, 67–78 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Wculek, S. K. & Malanchi, I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 528, 413–417 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Acharyya, S. et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 150, 165–178 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Wang, H. et al. The osteogenic niche promotes early-stage bone colonization of disseminated breast cancer cells. Cancer Cell 27, 193–210 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Zheng, H. et al. therapeutic antibody targeting tumor- and osteoblastic niche-derived jagged1 sensitizes bone metastasis to chemotherapy. Cancer Cell 32, 731–747 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  52. Chen, Q. et al. Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 533, 493–498 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Fong, M. Y. et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 17, 183–194 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Loo, J. M. et al. Extracellular metabolic energetics can promote cancer progression. Cell 160, 393–406 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Neman, J. et al. Human breast cancer metastases to the brain display GABAergic properties in the neural niche. Proc. Natl Acad. Sci. USA 111, 984–989 (2014).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  56. Egeblad, M., Rasch, M. G. & Weaver, V. M. Dynamic interplay between the collagen scaffold and tumor evolution. Curr. Opin. Cell Biol. 22, 697–706 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Vennin, C. et al. Transient tissue priming via ROCK inhibition uncouples pancreatic cancer progression, sensitivity to chemotherapy, and metastasis. Sci. Transl Med. 9, eaai8504 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Wan, L., Pantel, K. & Kang, Y. Tumor metastasis: moving new biological insights into the clinic. Nat. Med. 19, 1450–1464 (2013).

    Article  PubMed  CAS  Google Scholar 

  59. Schrader, J. et al. Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells. Hepatology 53, 1192–1205 (2011).

    Article  PubMed  CAS  Google Scholar 

  60. Shibue, T., Brooks, M. W., Inan, M. F., Reinhardt, F. & Weinberg, R. A. The outgrowth of micrometastases is enabled by the formation of filopodium-like protrusions. Cancer Discov. 2, 706–721 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Lambert, A. W., Pattabiraman, D. R. & Weinberg, R. A. Emerging biological principles of metastasis. Cell 168, 670–691 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Giancotti, F. G. Mechanisms governing metastatic dormancy and reactivation. Cell 155, 750–764 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Lu, X. et al. VCAM-1 promotes osteolytic expansion of indolent bone micrometastasis of breast cancer by engaging alpha4beta1-positive osteoclast progenitors. Cancer Cell 20, 701–714 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Sosa, M. S., Bragado, P. & Aguirre-Ghiso, J. A. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat. Rev. Cancer 14, 611–622 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Bragado, P. et al. TGF-β2 dictates disseminated tumour cell fate in target organs through TGF-β-RIII and p38α/β signalling. Nat. Cell Biol. 15, 1351–1361 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Ghajar, C. M. Metastasis prevention by targeting the dormant niche. Nat. Rev. Cancer 15, 238–247 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Varga, J. & Greten, F. R. Cell plasticity in epithelial homeostasis and tumorigenesis. Nat. Cell Biol. 19, 1133–1141 (2017).

    Article  PubMed  CAS  Google Scholar 

  68. Mosteiro, L. et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science 354, aaf4445 (2016).

    Article  PubMed  CAS  Google Scholar 

  69. Batlle, E. & Clevers, H. Cancer stem cells revisited. Nat. Med. 23, 1124–1134 (2017).

    Article  PubMed  CAS  Google Scholar 

  70. Gupta, P. B. et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146, 633–644 (2011).

    Article  PubMed  CAS  Google Scholar 

  71. Hendrix, M. J. et al. Reprogramming metastatic tumour cells with embryonic microenvironments. Nat. Rev. Cancer 7, 246–255 (2007).

    Article  PubMed  CAS  Google Scholar 

  72. Pascual, G. et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 541, 41–45 (2017).

    Article  PubMed  CAS  Google Scholar 

  73. de Sousa e Melo, F. et al. A distinct role for Lgr5+ stem cells in primary and metastatic colon cancer. Nature 543, 676–680 (2017).

    Article  PubMed  CAS  Google Scholar 

  74. Oskarsson, T. et al. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat. Med. 17, 867–874 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Yu, H., Lee, H., Herrmann, A., Buettner, R. & Jove, R. Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat. Rev. Cancer 14, 736–746 (2014).

    Article  PubMed  CAS  Google Scholar 

  76. Calon, A. et al. Dependency of colorectal cancer on a TGF-beta-driven program in stromal cells for metastasis initiation. Cancer Cell 22, 571–584 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Roorda, B. D., Elst, A., Boer, T. G., Kamps, W. A. & de Bont, E. S. Mesenchymal stem cells contribute to tumor cell proliferation by direct cell-cell contact interactions. Cancer Invest. 28, 526–534 (2010).

    Article  PubMed  CAS  Google Scholar 

  78. Cuiffo, B. G. et al. MSC-regulated microRNAs converge on the transcription factor FOXP2 and promote breast cancer metastasis. Cell Stem Cell 15, 762–774 (2014).

    Article  PubMed  CAS  Google Scholar 

  79. Ren, G., Esposito, M. & Kang, Y. Bone metastasis and the metastatic niche. J. Mol. Med. 93, 1203–1212 (2015).

    Article  PubMed  CAS  Google Scholar 

  80. Ridge, S. M., Sullivan, F. J. & Glynn, S. A. Mesenchymal stem cells: key players in cancer progression. Mol. Cancer 16, 31 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Celia-Terrassa, T. et al. Normal and cancerous mammary stem cells evade interferon-induced constraint through the miR-199a-LCOR axis. Nat. Cell Biol. 19, 711–723 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Pencheva, N. et al. Convergent multi-miRNA targeting of ApoE drives LRP1/LRP8-dependent melanoma metastasis and angiogenesis. Cell 151, 1068–1082 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Xing, F. et al. Reactive astrocytes promote the metastatic growth of breast cancer stem-like cells by activating Notch signalling in brain. EMBO Mol. Med. 5, 384–396 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Gao, H. et al. The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites. Cell 150, 764–779 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Malladi, S. et al. Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell 165, 45–60 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Mani, S. A. et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Korpal, M. et al. Direct targeting of Sec23a by miR-200s influences cancer cell secretome and promotes metastatic colonization. Nat. Med. 17, 1101–1108 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Ocana, O. H. et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724 (2012).

    Article  PubMed  CAS  Google Scholar 

  90. Tsai, J. H., Donaher, J. L., Murphy, D. A., Chau, S. & Yang, J. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell 22, 725–736 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Celia-Terrassa, T. et al. Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells. J. Clin. Invest. 122, 1849–1868 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Tran, H. D. et al. Transient SNAIL1 expression is necessary for metastatic competence in breast cancer. Cancer Res. 74, 6330–6340 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Fischer, K. R. et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 527, 472–476 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Zheng, X. et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 527, 525–530 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Nieto, M. A., Huang, R. Y., Jackson, R. A. & Thiery, J. P. Emt: 2016. Cell 166, 21–45 (2016).

    Article  PubMed  CAS  Google Scholar 

  96. Pastushenko, I. et al. Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018).

    Article  PubMed  CAS  Google Scholar 

  97. Kalluri, R. & Weinberg, R. A. The basics of epithelial-mesenchymal transition. J. Clin. Invest. 119, 1420–1428 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Lopez-Novoa, J. M. & Nieto, M. A. Inflammation and EMT: an alliance towards organ fibrosis and cancer progression. EMBO Mol. Med. 1, 303–314 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Wei, S. C. et al. Matrix stiffness drives epithelial-mesenchymal transition and tumour metastasis through a TWIST1–G3BP2 mechanotransduction pathway. Nat. Cell Biol. 17, 678–688 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Thiery, J. P., Acloque, H., Huang, R. Y. & Nieto, M. A. Epithelial-mesenchymal transitions in development and disease. Cell 139, 871–890 (2009).

    Article  PubMed  CAS  Google Scholar 

  101. Massague, J. TGF-β in cancer. Cell 134, 215–230 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Lawson, D. A. et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature 526, 131–135 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Gao, D. et al. Myeloid progenitor cells in the premetastatic lung promote metastases by inducing mesenchymal to epithelial transition. Cancer Res. 72, 1384–1394 (2012).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  104. Ouzounova, M. et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat. Commun. 8, 14979 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Salvador, F. et al. Lysyl oxidase-like protein LOXL2 promotes lung metastasis of breast cancer. Cancer Res. 77, 5846–5859 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Del Pozo Martin, Y. et al. Mesenchymal cancer cell-stroma crosstalk promotes niche activation, epithelial reversion, and metastatic colonization. Cell Rep. 13, 2456–2469 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Beerling, E. et al. Plasticity between epithelial and mesenchymal states unlinks EMT from metastasis-enhancing stem cell capacity. Cell Rep. 14, 2281–2288 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Kitamura, T., Qian, B. Z. & Pollard, J. W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 15, 73–86 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Condamine, T., Ramachandran, I., Youn, J. I. & Gabrilovich, D. I. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu. Rev. Med. 66, 97–110 (2015).

    Article  PubMed  CAS  Google Scholar 

  110. Krall, J. A. et al. The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci. Transl Med. 10, eean3464 (2018).

    Article  Google Scholar 

  111. Zhuang, X. et al. Differential effects on lung and bone metastasis of breast cancer by Wnt signalling inhibitor DKK1. Nat. Cell Biol. 19, 1274–1285 (2017).

    Article  PubMed  CAS  Google Scholar 

  112. Wieland, E. et al. Endothelial Notch1 activity facilitates metastasis. Cancer Cell 31, 355–367 (2017).

    Article  PubMed  CAS  Google Scholar 

  113. Branzk, N. & Papayannopoulos, V. Molecular mechanisms regulating NETosis in infection and disease. Semin. Immunopathol. 35, 513–530 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Park, J. et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl Med. 8, 361ra138 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Marvel, D. & Gabrilovich, D. I. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J. Clin. Invest. 125, 3356–3364 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Liu, Y. et al. Premetastatic soil and prevention of breast cancer brain metastasis. Neuro. Oncol. 15, 891–903 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Sangaletti, S. et al. Osteopontin shapes immunosuppression in the metastatic niche. Cancer Res. 74, 4706–4719 (2014).

    Article  PubMed  CAS  Google Scholar 

  118. Welte, T. et al. Oncogenic mTOR signalling recruits myeloid-derived suppressor cells to promote tumour initiation. Nat. Cell Biol. 18, 632–644 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Steele, C. W. et al. CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell 29, 832–845 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Smith, H. A. & Kang, Y. The metastasis-promoting roles of tumor-associated immune cells. J. Mol. Med. 91, 411–429 (2013).

    Article  PubMed  CAS  Google Scholar 

  121. Viguier, M. et al. Foxp3 expressing CD4+CD25(high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. J. Immunol. 173, 1444–1453 (2004).

    Article  PubMed  CAS  Google Scholar 

  122. Dunn, G. P., Old, L. J. & Schreiber, R. D. The immunobiology of cancer immunosurveillance and immunoediting. Immunity 21, 137–148 (2004).

    Article  PubMed  CAS  Google Scholar 

  123. Giraldo, N. A. et al. The immune contexture of primary and metastatic human tumours. Curr. Opin. Immunol. 27, 8–15 (2014).

    Article  PubMed  CAS  Google Scholar 

  124. Eyles, J. et al. Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J. Clin. Invest. 120, 2030–2039 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Lopez-Soto, A., Gonzalez, S., Smyth, M. J. & Galluzzi, L. Control of metastasis by NK cells. Cancer Cell 32, 135–154 (2017).

    Article  PubMed  CAS  Google Scholar 

  126. Schreiber, R. D., Old, L. J. & Smyth, M. J. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331, 1565–1570 (2011).

    Article  PubMed  CAS  Google Scholar 

  127. Paolino, M. et al. The E3 ligase Cbl-b and TAM receptors regulate cancer metastasis via natural killer cells. Nature 507, 508–512 (2014).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  128. Bidwell, B. N. et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 18, 1224–1231 (2012).

    Article  PubMed  CAS  Google Scholar 

  129. Payne, K. K. et al. Tumor-reactive immune cells protect against metastatic tumor and induce immunoediting of indolent but not quiescent tumor cells. J. Leukoc. Biol. 100, 625–635 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Kim, R., Emi, M. & Tanabe, K. Cancer immunoediting from immune surveillance to immune escape. Immunology 121, 1–14 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Rhim, A. D. et al. EMT and dissemination precede pancreatic tumor formation. Cell 148, 349–361 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Harper, K. L. et al. Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Nature 540, 588–592 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Hosseini, H. et al. Early dissemination seeds metastasis in breast cancer. Nature 540, 552–558 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Muller, M. et al. EblacZ tumor dormancy in bone marrow and lymph nodes: active control of proliferating tumor cells by CD8+ immune T cells. Cancer Res. 58, 5439–5446 (1998).

    PubMed  CAS  Google Scholar 

  135. Aguirre-Ghiso, J. A. Models, mechanisms and clinical evidence for cancer dormancy. Nat. Rev. Cancer 7, 834–846 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Davies, L. C., Jenkins, S. J., Allen, J. E. & Taylor, P. R. Tissue-resident macrophages. Nat. Immunol. 14, 986–995 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Fan, X. & Rudensky, A. Y. Hallmarks of tissue-resident lymphocytes. Cell 164, 1198–1211 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Jimenez-Sanchez, A. et al. Heterogeneous tumor-immune microenvironments among differentially growing metastases in an ovarian cancer patient. Cell 170, 927–938 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Smith, H. A. & Kang, Y. Determinants of organotropic metastasis. Annu. Rev. Cancer Biol. 1, 403–423 (2017).

    Article  Google Scholar 

  140. Wu, J. B. et al. MAOA-dependent activation of Shh-IL6-RANKL signaling network promotes prostate cancer metastasis by engaging tumor-stromal cell interactions. Cancer Cell 31, 368–382 (2017).

    Article  PubMed  CAS  Google Scholar 

  141. Nilsson, M., Adamo, H., Bergh, A. & Halin Bergstrom, S. Inhibition of lysyl oxidase and lysyl oxidase-like enzymes has tumour-promoting and tumour-suppressing roles in experimental prostate cancer. Sci. Rep. 6, 19608 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Shay, G., Lynch, C. C. & Fingleton, B. Moving targets: Emerging roles for MMPs in cancer progression and metastasis. Matrix Biol. 44–46, 200–206 (2015).

    Article  PubMed  CAS  Google Scholar 

  143. Shen, S. et al. Vascular endothelial growth factor enhances cancer cell adhesion to microvascular endothelium in vivo. Exp. Physiol. 95, 369–379 (2010).

    Article  PubMed  CAS  Google Scholar 

  144. Germano, G. et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 23, 249–262 (2013).

    Article  PubMed  CAS  Google Scholar 

  145. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Schachter, J. et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet 390, 1853–1862 (2017).

    Article  PubMed  CAS  Google Scholar 

  147. Bullman, S. et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 358, 1443–1448 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  148. Aguado, B. A., Bushnell, G. G., Rao, S. S., Jeruss, J. S. & Shea, L. D. Engineering the pre-metastatic niche. Nat. Biomed. Eng. 1, 0077 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  149. Pontes-Quero, S. et al. Dual ifgMosaic: a versatile method for multispectral and combinatorial mosaic gene-function analysis. Cell 170, 800–814 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of our laboratories for helpful discussions, and in particular, D. Liu for critical reading of the manuscript. We also apologize to the many investigators whose important studies could not be cited directly here owing to space limitations. The work was supported by a Susan G. Komen Fellowship (PDF15332075) and from Instituto de Salud Carlos III-FSE (MS17/00037) to T. C-T, and grants from the Brewster Foundation, the Breast Cancer Research Foundation, Department of Defense (BC123187), and the National Institutes of Health (R01CA141062) to Y.K.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yibin Kang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Celià-Terrassa, T., Kang, Y. Metastatic niche functions and therapeutic opportunities. Nat Cell Biol 20, 868–877 (2018). https://doi.org/10.1038/s41556-018-0145-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0145-9

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