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:

Targeting metastatic cancer

Abstract

Despite recent therapeutic advances in cancer treatment, metastasis remains the principal cause of cancer death. Recent work has uncovered the unique biology of metastasis-initiating cells that results in tumor growth in distant organs, evasion of immune surveillance and co-option of metastatic microenvironments. Here we review recent progress that is enabling therapeutic advances in treating both micro- and macrometastases. Such insights were gained from cancer sequencing, mechanistic studies and clinical trials, including of immunotherapy. These studies reveal both the origins and nature of metastases and identify new opportunities for developing more effective strategies to target metastatic relapse and improve patient outcomes.

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: Steps, biological functions and cancer cell vulnerabilities in the metastasis cascade.
Fig. 2: Model of metastasis subverting normal regenerative processes.
Fig. 3: Classic and new opportunities for the treatment of metastatic cancer.

Similar content being viewed by others

References

  1. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 70, 7–30 (2020).

    Article  PubMed  Google Scholar 

  2. Caswell-Jin, J. L. et al. Change in survival in metastatic breast cancer with treatment advances: meta-analysis and systematic review. JNCI Cancer Spectr. 2, pky062 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  3. Howlader, N. et al. (eds.) in SEER Cancer Statistics Review, 1975–2016 1423–1437 (National Cancer Institute, 2019).

  4. May, M. Twenty-five ways clinical trials have changed in the last 25 years. Nat. Med. 25, 2–5 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Hu, Z. & Curtis, C. Looking backward in time to define the chronology of metastasis. Nat. Commun. 11, 3213 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Chambers, A. F., Groom, A. C. & MacDonald, I. C. Dissemination and growth of cancer cells in metastatic sites. Nat. Rev. Cancer 2, 563–572 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  10. Massagué, J. & Obenauf, A. C. Metastatic colonization by circulating tumour cells. Nature 529, 298–306 (2016).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  11. Quintanal-Villalonga, A. et al. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat. Rev. Clin. Oncol. 17, 360–371 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  12. Laughney, A. M. et al. Regenerative lineages and immune-mediated pruning in lung cancer metastasis. Nat. Med. 26, 259–269 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Fumagalli, A. O. et al. Plasticity of Lgr5-negative cancer cells drives metastasis in colorectal cancer. Cell Stem Cell 26, 569–578 (2020).

  14. Biswas, D. et al. A clonal expression biomarker associates with lung cancer mortality. Nat. Med. 25, 1540–1548 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Obenauf, A. C. & Massagué, J. Surviving at a distance: organ-specific metastasis. Trends Cancer 1, 76–91 (2015).

    Article  PubMed Central  PubMed  Google Scholar 

  16. Esposito, M., Guise, T. & Kang, Y. The biology of bone metastasis. Cold Spring Harb. Perspect. Med. https://doi.org/10.1101/cshperspect.a031252 (2018).

  17. Boire, A., Brastianos, P. K., Garzia, L. & Valiente, M. Brain metastasis. Nat. Rev. Cancer 20, 4–11 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Celià-Terrassa, T. & Kang, Y. Metastatic niche functions and therapeutic opportunities. Nat. Cell Biol. 20, 868–877 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Turajlic, S. & Swanton, C. Metastasis as an evolutionary process. Science 352, 169–175 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Reiter, J. G. et al. An analysis of genetic heterogeneity in untreated cancers. Nat. Rev. Cancer 19, 639–650 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Razavi, P. et al. The genomic landscape of endocrine-resistant advanced breast cancers. Cancer Cell 34, 427–438 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Reiter, J. G. et al. Minimal functional driver gene heterogeneity among untreated metastases. Science 361, 1033–1037 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Sanchez-Vega, F. et al. Oncogenic signaling pathways in The Cancer Genome Atlas. Cell 173, 321–337 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Hu, Z., Li, Z., Ma, Z. & Curtis, C. Multi-cancer analysis of clonality and the timing of systemic spread in paired primary tumors and metastases. Nat. Genet. 52, 701–708 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Jacob, L. S. et al. Metastatic competence can emerge with selection of preexisting oncogenic alleles without a need of new mutations. Cancer Res. 75, 3713–3719 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Denny, S. K. et al. Nfib promotes metastasis through a widespread increase in chromatin accessibility. Cell 166, 328–342 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Roe, J. S. et al. Enhancer reprogramming promotes pancreatic cancer metastasis. Cell 170, 875–888 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Vanharanta, S. et al. Epigenetic expansion of VHL–HIF signal output drives multiorgan metastasis in renal cancer. Nat. Med. 19, 50–56 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Tavazoie, S. F. et al. Endogenous human microRNAs that suppress breast cancer metastasis. Nature 451, 147–152 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Gomes, A. P. et al. Dynamic incorporation of histone H3 variants into chromatin is essential for acquisition of aggressive traits and metastatic colonization. Cancer Cell 36, 402–417 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. McDonald, O. G. et al. Epigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasis. Nat. Genet. 49, 367–376 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  32. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  34. Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648 (1994).

    Article  CAS  PubMed  Google Scholar 

  35. Bonnet, D. & Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 3, 730–737 (1997).

    Article  CAS  PubMed  Google Scholar 

  36. Shimokawa, M. et al. Visualization and targeting of LGR5+ human colon cancer stem cells. Nature 545, 187–192 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Barker, N. et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Visvader, J. E. Cells of origin in cancer. Nature 469, 314–322 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Ganesh, K. et al. L1CAM defines the regenerative origin of metastasis-initiating cells in colorectal cancer. Nat. Cancer 1, 28–45 (2020).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  41. Altevogt, P., Doberstein, K. & Fogel, M. L1CAM in human cancer. Int. J. Cancer 138, 1565–1576 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Er, E. E. et al. Pericyte-like spreading by disseminated cancer cells activates YAP and MRTF for metastatic colonization. Nat. Cell Biol. 20, 966–978 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  45. Yang, J. et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 21, 341–352 (2020).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  47. Shibue, T. & Weinberg, R. A. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 14, 611–629 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  48. 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  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  50. Su, J. et al. TGF-β orchestrates fibrogenic and developmental EMTs via the RAS effector RREB1. Nature 577, 566–571 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Rickman, D. S., Beltran, H., Demichelis, F. & Rubin, M. A. Biology and evolution of poorly differentiated neuroendocrine tumors. Nat. Med. 23, 1–10 (2017).

    Article  CAS  PubMed  Google Scholar 

  52. Yuan, S., Norgard, R. J. & Stanger, B. Z. Cellular plasticity in cancer. Cancer Discov. 9, 837–851 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Boumahdi, S. & de Sauvage, F. J. The great escape: tumour cell plasticity in resistance to targeted therapy. Nat. Rev. Drug Discov. 19, 39–56 (2020).

    Article  CAS  PubMed  Google Scholar 

  54. Lugassy, C., Kleinman, H. K., Vermeulen, P. B. & Barnhill, R. L. Angiotropism, pericytic mimicry and extravascular migratory metastasis: an embryogenesis-derived program of tumor spread. Angiogenesis 23, 27–41 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Kuol, N., Stojanovska, L., Apostolopoulos, V. & Nurgali, K. Role of the nervous system in cancer metastasis. J. Exp. Clin. Cancer Res. 37, 5 (2018).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  56. Yu, M. et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 339, 580–584 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Gkountela, S. et al. Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding. Cell 176, 98–112 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. Pantel, K. & Speicher, M. R. The biology of circulating tumor cells. Oncogene 35, 1216–1224 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Micalizzi, D. S., Maheswaran, S. & Haber, D. A. A conduit to metastasis: circulating tumor cell biology. Genes Dev. 31, 1827–1840 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  60. Fidler, I. J. The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis. Eur. J. Cancer 9, 223–227 (1973).

    Article  CAS  PubMed  Google Scholar 

  61. Aceto, N., Toner, M., Maheswaran, S. & Haber, D. A. En route to metastasis: circulating tumor cell clusters and epithelial-to-mesenchymal transition. Trends Cancer 1, 44–52 (2015).

    Article  PubMed  Google Scholar 

  62. Chemi, F. et al. Pulmonary venous circulating tumor cell dissemination before tumor resection and disease relapse. Nat. Med. 25, 1534–1539 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Heller, G. et al. Circulating tumor cell number as a response measure of prolonged survival for metastatic castration-resistant prostate cancer: a comparison with prostate-specific antigen across five randomized phase III clinical trials. J. Clin. Oncol. 36, 572–580 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Alix-Panabieres, C., Schwarzenbach, H. & Pantel, K. Circulating tumor cells and circulating tumor DNA. Annu. Rev. Med. 63, 199–215 (2012).

    Article  CAS  PubMed  Google Scholar 

  65. Cristofanilli, M. et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): international expert consensus paper. Crit. Rev. Oncol. Hematol. 134, 39–45 (2019).

    Article  PubMed  Google Scholar 

  66. Pantel, K., Hille, C. & Scher, H. I. Circulating tumor cells in prostate cancer: from discovery to clinical utility. Clin. Chem. 65, 87–99 (2019).

    Article  CAS  PubMed  Google Scholar 

  67. Parikh, A. R. et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 25, 1415–1421 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  68. 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  CAS  PubMed Central  PubMed  Google Scholar 

  69. Phan, T. G. & Croucher, P. I. The dormant cancer cell life cycle. Nat. Rev. Cancer 20, 398–411 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Carlson, P. et al. Targeting the perivascular niche sensitizes disseminated tumour cells to chemotherapy. Nat. Cell Biol. 21, 238–250 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  73. David, C. J. & Massagué, J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat. Rev. Mol. Cell Biol. 19, 419–435 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  74. Bragado, P. et al. TGF-β2 dictates disseminated tumour cell fate in target organs through TGF-β-RIII and p38ɑ/β signalling. Nat. Cell Biol. 15, 1351–1361 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  75. Prunier, C., Baker, D., Ten Dijke, P. & Ritsma, L. TGF-β family signaling pathways in cellular dormancy. Trends Cancer 5, 66–78 (2019).

    Article  CAS  PubMed  Google Scholar 

  76. Pommier, A. et al. Unresolved endoplasmic reticulum stress engenders immune-resistant, latent pancreatic cancer metastases. Science https://doi.org/10.1126/science.aao4908 (2018).

  77. Pantel, K. et al. Frequent down-regulation of major histocompatibility class I antigen expression on individual micrometastatic carcinoma cells. Cancer Res. 51, 4712–4715 (1991).

    CAS  PubMed  Google Scholar 

  78. Koebel, C. M. et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450, 903–907 (2007).

    Article  CAS  PubMed  Google Scholar 

  79. Eyob, H. et al. Inhibition of Ron kinase blocks conversion of micrometastases to overt metastases by boosting antitumor immunity. Cancer Discov. 3, 751–760 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  80. Ali, F. R. & Lear, J. T. Melanoma in organ transplant recipients: incidence, outcomes and management considerations. J. Skin Cancer 2012, 404615 (2012).

    Article  PubMed Central  PubMed  Google Scholar 

  81. Tikhonova, A. N., Lasry, A., Austin, R. & Aifantis, I. Cell-by-cell deconstruction of stem cell niches. Cell Stem Cell 27, 19–34 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  83. 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  CAS  PubMed Central  PubMed  Google Scholar 

  84. Mohme, M., Riethdorf, S. & Pantel, K. Circulating and disseminated tumour cells—mechanisms of immune surveillance and escape. Nat. Rev. Clin. Oncol. 14, 155–167 (2017).

    Article  CAS  PubMed  Google Scholar 

  85. Garner, H. & de Visser, K. E. Immune crosstalk in cancer progression and metastatic spread: a complex conversation. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-019-0271-z (2020).

  86. Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  87. 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  CAS  PubMed Central  PubMed  Google Scholar 

  88. 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  CAS  PubMed  Google Scholar 

  89. Van den Eynde, M. et al. The link between the multiverse of immune microenvironments in metastases and the survival of colorectal cancer patients. Cancer Cell 34, 1012–1026 (2018).

    Article  PubMed  CAS  Google Scholar 

  90. Kang, Y. et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003).

    Article  CAS  PubMed  Google Scholar 

  91. 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  CAS  PubMed Central  PubMed  Google Scholar 

  92. Zhang, J. et al. Osteoprotegerin inhibits prostate cancer-induced osteoclastogenesis and prevents prostate tumor growth in the bone. J. Clin. Invest. 107, 1235–1244 (2001).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  93. Chen, Q., Zhang, X. H. & Massagué, 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  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  95. Albrengues, J. et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science https://doi.org/10.1126/science.aao4227 (2018).

  96. Teijeira, A. et al. CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity 52, 856–871 (2020).

    Article  CAS  PubMed  Google Scholar 

  97. Ombrato, L. et al. Metastatic-niche labelling reveals parenchymal cells with stem features. Nature 572, 603–608 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  99. Sevenich, L. et al. Analysis of tumour- and stroma-supplied proteolytic networks reveals a brain-metastasis-promoting role for cathepsin S. Nat. Cell Biol. 16, 876–888 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  100. Zeng, Q. et al. Synaptic proximity enables NMDAR signalling to promote brain metastasis. Nature 573, 526–531 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  103. Fontebasso, Y. & Dubinett, S. M. Drug development for metastasis prevention. Crit. Rev. Oncog. 20, 449–473 (2015).

    Article  PubMed Central  PubMed  Google Scholar 

  104. Risson, E., Nobre, A. R., Maguer-Satta, V. & Aguirre-Ghiso, J. A. The current paradigm and challenges ahead for the dormancy of disseminated tumor cells. Nat. Cancer 1, 672–680 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Parsons, S., Maldonado, E. B. & Prasad, V. Comparison of drugs used for adjuvant and metastatic therapy of colon, breast, and non-small cell lung cancers. JAMA Netw. Open 3, e202488 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  106. Steeg, P. S. Perspective: The right trials. Nature 485, S58–59 (2012).

    Article  CAS  PubMed  Google Scholar 

  107. Vasan, N., Baselga, J. & Hyman, D. M. A view on drug resistance in cancer. Nature 575, 299–309 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Lito, P., Rosen, N. & Solit, D. B. Tumor adaptation and resistance to RAF inhibitors. Nat. Med. 19, 1401–1409 (2013).

    Article  CAS  PubMed  Google Scholar 

  109. Sabnis, A. J. & Bivona, T. G. Principles of resistance to targeted cancer therapy: lessons from basic and translational cancer biology. Trends Mol. Med. 25, 185–197 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  110. Wu, Y.-L. et al. CTONG1104: adjuvant gefitinib versus chemotherapy for resected N1-N2 NSCLC with EGFR mutation—final overall survival analysis of the randomized phase III trial 1 analysis of the randomized phase III trial. J. Clin. Oncol. 38, 9005 (2020).

    Article  Google Scholar 

  111. Jassem, J. Adjuvant EGFR tyrosine kinase inhibitors in EGFR-mutant non-small cell lung cancer: still an investigational approach. Transl. Lung Cancer Res. 8, S387–S390 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  112. Mok, T. S. et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N. Engl. J. Med. 376, 629–640 (2017).

    Article  CAS  PubMed  Google Scholar 

  113. Herbst, R. S. et al. Osimertinib as adjuvant therapy in patients (pts) with stage IB–IIIA EGFR mutation positive (EGFRm) NSCLC after complete tumor resection: ADAURA. J. Clin. Oncol. 38, abstract LBA5 (2020).

  114. Soria, J. C. et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N. Engl. J. Med. 378, 113–125 (2018).

    Article  CAS  PubMed  Google Scholar 

  115. Mateo, J. et al. A decade of clinical development of PARP inhibitors in perspective. Ann. Oncol. 30, 1437–1447 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  116. Farmer, H. et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005).

    Article  CAS  PubMed  Google Scholar 

  117. Spring, L. M. et al. Cyclin-dependent kinase 4 and 6 inhibitors for hormone receptor-positive breast cancer: past, present, and future. Lancet 395, 817–827 (2020).

    Article  CAS  PubMed  Google Scholar 

  118. Paget, S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 8, 98–101 (1989).

    CAS  PubMed  Google Scholar 

  119. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  120. Folkman, J. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. 285, 1182–1186 (1971).

    Article  CAS  PubMed  Google Scholar 

  121. Haibe, Y. et al. Resistance mechanisms to anti-angiogenic therapies in cancer. Front. Oncol. 10, 221 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  122. Iheanacho, K. & Vaishampayan, U. Perioperative approaches to kidney cancer. Clin. Adv. Hematol. Oncol. 18, 56–65 (2020).

    PubMed  Google Scholar 

  123. Allegra, C. J. et al. Bevacizumab in stage II–III colon cancer: 5-year update of the National Surgical Adjuvant Breast and Bowel Project C-08 trial. J. Clin. Oncol. 31, 359–364 (2013).

    Article  CAS  PubMed  Google Scholar 

  124. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  125. Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381, 1535–1546 (2019).

    Article  CAS  PubMed  Google Scholar 

  126. Haslam, A., Gill, J. & Prasad, V. Estimation of the percentage of US patients with cancer who are eligible for immune checkpoint inhibitor drugs. JAMA Netw. Open 3, e200423 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  127. Hellmann, M. D. et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. Lancet Oncol. 18, 31–41 (2017).

    Article  CAS  PubMed  Google Scholar 

  128. Motzer, R. J. et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N. Engl. J. Med. 378, 1277–1290 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  129. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  130. Long, G. V. et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol. 19, 672–681 (2018).

    Article  CAS  PubMed  Google Scholar 

  131. Schumacher, T. N., Scheper, W. & Kvistborg, P. Cancer neoantigens. Annu. Rev. Immunol. 37, 173–200 (2019).

    Article  CAS  PubMed  Google Scholar 

  132. Hellmann, M. D. et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N. Engl. J. Med. 378, 2093–2104 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  133. Marabelle, A. et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 Study. J. Clin. Oncol. 38, 1–10 (2020).

    Article  CAS  PubMed  Google Scholar 

  134. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  135. Rizvi, N. A. et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  136. Tauriello, D. V. F. et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 554, 538–543 (2018).

    Article  CAS  PubMed  Google Scholar 

  137. Mariathasan, S. et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  138. Makker, V. et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: an interim analysis of a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 20, 711–718 (2019).

    Article  CAS  PubMed  Google Scholar 

  139. Rini, B. I. et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N. Engl. J. Med. 380, 1116–1127 (2019).

    Article  CAS  PubMed  Google Scholar 

  140. Fukuoka, S. et al. Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J. Clin. Oncol. 38, 2053–2061 (2020).

    Article  CAS  PubMed  Google Scholar 

  141. Eggermont, A. M. M. et al. Adjuvant pembrolizumab versus placebo in resected stage III melanoma. N. Engl. J. Med. 378, 1789–1801 (2018).

    Article  CAS  PubMed  Google Scholar 

  142. Weber, J. et al. Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N. Engl. J. Med. 377, 1824–1835 (2017).

    Article  CAS  PubMed  Google Scholar 

  143. Eggermont, A. M. et al. Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy. N. Engl. J. Med. 375, 1845–1855 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  144. Zimmer, L. et al. Adjuvant nivolumab plus ipilimumab or nivolumab monotherapy versus placebo in patients with resected stage IV melanoma with no evidence of disease (IMMUNED): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 395, 1558–1568 (2020).

    Article  CAS  PubMed  Google Scholar 

  145. Liu, J. et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discov. 6, 1382–1399 (2016).

    Article  CAS  PubMed  Google Scholar 

  146. Versluis, J. M., Long, G. V. & Blank, C. U. Learning from clinical trials of neoadjuvant checkpoint blockade. Nat. Med. 26, 475–484 (2020).

    Article  CAS  PubMed  Google Scholar 

  147. Blank, C. U. et al. Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat. Med. 24, 1655–1661 (2018).

    Article  CAS  PubMed  Google Scholar 

  148. Amaria, R. N. et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat. Med. 24, 1649–1654 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  149. Forde, P. M. et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 378, 1976–1986 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  150. Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711–723 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  151. Creasy, J. M. et al. Actual 10-year survival after hepatic resection of colorectal liver metastases: what factors preclude cure? Surgery 163, 1238–1244 (2018).

    Article  PubMed  Google Scholar 

  152. Clark, M. E. & Smith, R. R. Liver-directed therapies in metastatic colorectal cancer. J. Gastrointest. Oncol. 5, 374–387 (2014).

    PubMed Central  PubMed  Google Scholar 

  153. Gomez, D. R. et al. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J. Clin. Oncol. 37, 1558–1565 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  154. Palma, D. A. et al. Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): a randomised, phase 2, open-label trial. Lancet 393, 2051–2058 (2019).

    Article  PubMed  Google Scholar 

  155. Parker, C. et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N. Engl. J. Med. 369, 213–223 (2013).

    Article  CAS  PubMed  Google Scholar 

  156. Early Breast Cancer Trialists’ Collaborative Group. Adjuvant bisphosphonate treatment in early breast cancer: meta-analyses of individual patient data from randomised trials. Lancet 386, 1353–1361 (2015).

    Article  CAS  Google Scholar 

  157. O’Carrigan, B. et al. Bisphosphonates and other bone agents for breast cancer. Cochrane Database of Systematic Reviews https://doi.org/10.1002/14651858.CD003474.pub4 (2017).

  158. Lin, X. & DeAngelis, L. M. Treatment of brain metastases. J. Clin. Oncol. 33, 3475–3484 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  159. Auperin, A. et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. Prophylactic Cranial Irradiation Overview Collaborative Group. N. Engl. J. Med. 341, 476–484 (1999).

    Article  CAS  PubMed  Google Scholar 

  160. Boire, A. et al. Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis. Cell 168, 1101–1113 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  161. Haro, M. A., Dyevoich, A. M., Phipps, J. P. & Haas, K. M. Activation of B-1 cells promotes tumor cell killing in the peritoneal cavity. Cancer Res. 79, 159–170 (2019).

    Article  CAS  PubMed  Google Scholar 

  162. Priego, N. et al. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat. Med. 24, 1024–1035 (2018).

    Article  CAS  PubMed  Google Scholar 

  163. Chi, Y. et al. Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis. Science 369, 276–282 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Suva, M. L. & Tirosh, I. Single-cell RNA sequencing in cancer: lessons learned and emerging challenges. Mol. Cell 75, 7–12 (2019).

    Article  CAS  PubMed  Google Scholar 

  165. Rozenblatt-Rosen, O. et al. The Human Tumor Atlas Network: charting tumor transitions across space and time at single-cell resolution. Cell 181, 236–249 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  167. Rekhtman, N. et al. Next-generation sequencing of pulmonary large cell neuroendocrine carcinoma reveals small cell carcinoma-like and non-small cell carcinoma-like subsets. Clin. Cancer Res. 22, 3618–3629 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  168. Hamilton, K. et al. Prevalence and prognostic significance of neuroendocrine cells in esophageal adenocarcinoma. Mod. Pathol. 13, 475–481 (2000).

    Article  CAS  PubMed  Google Scholar 

  169. Adams, E. J. et al. FOXA1 mutations alter pioneering activity, differentiation and prostate cancer phenotypes. Nature 571, 408–412 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  170. Kaur, J., Daoud, A. & Eblen, S. T. Targeting chromatin remodeling for cancer therapy. Curr. Mol. Pharm. 12, 215–229 (2019).

    Article  CAS  Google Scholar 

  171. Sparano, J. A. et al. Clinical and genomic risk to guide the use of adjuvant therapy for breast cancer. N. Engl. J. Med. 380, 2395–2405 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  172. Sobrero, A. F. et al. Overall survival (OS) and long-term disease-free survival (DFS) of three versus six months of adjuvant (adj) oxaliplatin and fluoropyrimidine-based therapy for patients (pts) with stage III colon cancer (CC): final results from the IDEA (International Duration Evaluation of Adj chemotherapy) collaboration. J. Clin. Oncol. 38, 4004–4004 (2020).

    Article  Google Scholar 

  173. Grothey, A. et al. Duration of adjuvant chemotherapy for stage III colon cancer. N. Engl. J. Med. 378, 1177–1188 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  174. Coakley, M., Garcia-Murillas, I. & Turner, N. C. Molecular residual disease and adjuvant trial design in solid tumors. Clin. Cancer Res. 25, 6026–6034 (2019).

    Article  CAS  PubMed  Google Scholar 

  175. Pantel, K. & Alix-Panabières, C. Liquid biopsy and minimal residual disease—latest advances and implications for cure. Nat. Rev. Clin. Oncol. 16, 409–424 (2019).

    Article  CAS  PubMed  Google Scholar 

  176. Phallen, J. et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aan2415 (2017).

  177. Cohen, J. D. et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 359, 926–930 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  178. Razavi, P. et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat. Med. 25, 1928–1937 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  179. Helmink, B. A., Khan, M. A. W., Hermann, A., Gopalakrishnan, V. & Wargo, J. A. The microbiome, cancer, and cancer therapy. Nat. Med. 25, 377–388 (2019).

    Article  CAS  PubMed  Google Scholar 

  180. Geller, L. T. et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 357, 1156–1160 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  181. Nejman, D. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368, 973–980 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  183. Parhi, L. et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat. Commun. 11, 3259 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  184. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    Article  CAS  PubMed  Google Scholar 

  185. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

    Article  CAS  PubMed  Google Scholar 

  186. Forbes, N. S. et al. White paper on microbial anti-cancer therapy and prevention. J. Immunother. Cancer 6, 78 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  187. Tuveson, D. & Clevers, H. Cancer modeling meets human organoid technology. Science 364, 952–955 (2019).

    Article  CAS  PubMed  Google Scholar 

  188. Vlachogiannis, G. et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 359, 920–926 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  189. Ganesh, K. et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat. Med. 25, 1607–1614 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  190. Hapach, L. A., Mosier, J. A., Wang, W. & Reinhart-King, C. A. Engineered models to parse apart the metastatic cascade. NPJ Precis Oncol. 3, 20 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  191. Yuki, K., Cheng, N., Nakano, M. & Kuo, C. J. Organoid models of tumor immunology. Trends Immunol. https://doi.org/10.1016/j.it.2020.06.010 (2020).

  192. De La Rochere, P. et al. Humanized mice for the study of immuno-oncology. Trends Immunol. 39, 748–763 (2018).

    Article  CAS  Google Scholar 

  193. Priestley, P. et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature 575, 210–216 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  194. Balachandran, V. P. et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 551, 512–516 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  195. Iyer, G. et al. Genome sequencing identifies a basis for everolimus sensitivity. Science 338, 221 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  196. Doebele, R. C. et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1–2 trials. Lancet Oncol. 21, 271–282 (2020).

    Article  CAS  PubMed  Google Scholar 

  197. Prahallad, A. et al. Unresponsiveness of colon cancer to BRAFV600E inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012).

    Article  CAS  PubMed  Google Scholar 

  198. Dvorak, H. F. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N. Engl. J. Med. 315, 1650–1659 (1986).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors’ work in the subject area of this Review is supported by NIH grants R35CA252978 (J.M.), K08CA23021 (K.G.), P30-CA008748 (MSKCC) and the Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center at MSKCC (J.M.).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Karuna Ganesh or Joan Massagué.

Ethics declarations

Competing interests

J.M. owns stock of Scholar Rock, Inc. J.M. is an inventor on the following patents and patent applications: Assay for anti-metastatic agents (inventors: J.M. and L. Norton; no. 7,829,066; issued 9 November 2010), S100A8/A9 as a diagnostic marker and a therapeutic agent (inventor: J.M.; no. 2,831,593; granted 30 June 2018), Inhibiting cancer metastasis (inventors: J.M. and M. Valiente Cortes; no. 3,047,039; granted 31 July 2019), Methods for treating brain metastasis (inventors: J.M., Q. Chen and A. Boire; no. 10,413,522; issued 17 September 2019), Modulating permeability of the blood cerebrospinal fluid barrier (inventors: J.M. and A. A. Boire; application no. PCT/US2016/062880; published 18 November 2016). K.G. and J.M. are inventors on the following patent application: Treating metastatic cancer and model systems for metastatic disease (inventors: J.M., M. Valiente Cortes and K.G.; application no. PCT/US2017/045145; published 2 August 2017).

Additional information

Peer review information Hannah Stower was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Ganesh, K., Massagué, J. Targeting metastatic cancer. Nat Med 27, 34–44 (2021). https://doi.org/10.1038/s41591-020-01195-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-01195-4

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research