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:

Metastasis: from dissemination to organ-specific colonization

Key Points

  • Metastasis progression can be viewed as a stepwise sequence of events, which is mediated by different classes of metastasis genes.

  • For each type of cancer, the clinical course of these events occurs with distinct temporal kinetics and in unique organ sites.

  • The long latency period of certain tumour types suggests the further evolution or 'speciation' of malignant cells in the microenvironments of a particular organ. The acquisition of pro-metastatic functions earlier during primary tumour formation might enable other cancer subtypes to relapse more quickly.

  • The organ specificity of metastatic cells is determined by unique infiltrative and colonization functions required after their dissemination from a primary tumour.

  • New insights into the importance of latency and organ-specific colonization should be considered in the design of optimized therapeutic strategies.

Abstract

Metastasis to distant organs is an ominous feature of most malignant tumours but the natural history of this process varies in different cancers. The cellular origin, intrinsic properties of the tumour, tissue affinities and circulation patterns determine not only the sites of tumour spread, but also the temporal course and severity of metastasis to vital organs. Striking disparities in the natural progression of different cancers raise important questions about the evolution of metastatic traits, the genetic determinants of these properties and the mechanisms that lead to the selection of metastatic cells.

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: Basic steps of metastasis and hypothetical classes of metastasis genes.
Figure 2: Organ-specific barriers to metastatic infiltration.
Figure 3: Metastasis progression genes expressed in the primary tumour.
Figure 4: The temporal course of metastasis.
Figure 5: Metastatic speciation of latent disseminated tumour cells.

Similar content being viewed by others

References

  1. Christofori, G. New signals from the invasive front. Nature 441, 444–450 (2006).

    CAS  PubMed  Google Scholar 

  2. Gupta, G. P. & Massague, J. Cancer metastasis: building a framework. Cell 127, 679–695 (2006).

    CAS  PubMed  Google Scholar 

  3. Steeg, P. S. Tumor metastasis: mechanistic insights and clinical challenges. Nature Med. 12, 895–904 (2006).

    CAS  PubMed  Google Scholar 

  4. Fidler, I. J. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nature Rev. Cancer 3, 453–458 (2003).

    CAS  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  6. Yin, J. J. et al. TGF-β signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J. Clin. Invest. 103, 197–206 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Minn, A. J. et al. Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors. J. Clin. Invest. 115, 44–55 (2005). This paper shows that pleural effusion-derived metastatic cell populations are heterogeneous in their ability to colonize different organs, supporting the notion that various target organs impose different requirements on arriving tumour cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Minn, A. J. et al. Genes that mediate breast cancer metastasis to lung. Nature 436, 518–524 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Kang, Y. et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003). This paper shows that in vivo selection enriches for bone metastatic ability and identifies genetic mediators of bone metastatic colonization.

    CAS  PubMed  Google Scholar 

  10. Edlund, M., Sung, S. Y. & Chung, L. W. Modulation of prostate cancer growth in bone microenvironments. J. Cell. Biochem. 91, 686–705 (2004).

    CAS  PubMed  Google Scholar 

  11. Triozzi, P. L., Eng, C. & Singh, A. D. Targeted therapy for uveal melanoma. Cancer Treat. Rev. 34, 247–258 (2008).

    CAS  PubMed  Google Scholar 

  12. Hess, K. R. et al. Metastatic patterns in adenocarcinoma. Cancer 106, 1624–1633 (2006). A recent clinical study that reports the frequency of organ-specific relapse in 11 different types of adenocarcinomas from over 4,000 patients.

    PubMed  Google Scholar 

  13. Patanaphan, V., Salazar, O. M. & Risco, R. Breast cancer: metastatic patterns and their prognosis. South. Med. J. 81, 1109–1112 (1988).

    CAS  PubMed  Google Scholar 

  14. Schmidt-Kittler, O. et al. From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression. Proc. Natl Acad. Sci. USA 100, 7737–7742 (2003). This paper shows that disseminated tumour cells have different and fewer aberrations than their matched primary tumours, suggesting that dissemination is an early event during cancer development.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Karrison, T. G., Ferguson, D. J. & Meier, P. Dormancy of mammary carcinoma after mastectomy. J. Natl Cancer Inst. 91, 80–85 (1999).

    CAS  PubMed  Google Scholar 

  16. Feld, R., Rubinstein, L. V. & Weisenberger, T. H. Sites of recurrence in resected stage I non-small-cell lung cancer: a guide for future studies. J. Clin. Oncol. 2, 1352–1358 (1984).

    CAS  PubMed  Google Scholar 

  17. Hoffman, P. C., Mauer, A. M. & Vokes, E. E. Lung cancer. Lancet 355, 479–485 (2000).

    CAS  PubMed  Google Scholar 

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

    CAS  Google Scholar 

  19. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).

    CAS  PubMed  Google Scholar 

  20. Moody, S. E. et al. Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell 2, 451–461 (2002).

    CAS  PubMed  Google Scholar 

  21. Slamon, D. J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001).

    CAS  PubMed  Google Scholar 

  22. Minna, J. D., Kurie, J. M. & Jacks, T. A big step in the study of small cell lung cancer. Cancer Cell 4, 163–166 (2003).

    CAS  PubMed  Google Scholar 

  23. Klein, C. A. The systemic progression of human cancer: a focus on the individual disseminated cancer cell—the unit of selection. Adv. Cancer Res. 89, 35–67 (2003).

    CAS  PubMed  Google Scholar 

  24. Chiang, A. C. & Massagué, J. Molecular basis of metastasis. N. Engl. J. Med. 359, 2814–2823 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Nguyen, D. X. & Massagué, J. Genetic determinants of cancer metastasis. Nature Rev. Genet. 8, 341–352 (2007).

    CAS  PubMed  Google Scholar 

  26. Yang, J. & Weinberg, R. A. Epithelial–mesenchymal transition: at the crossroads of development and tumor metastasis. Dev. Cell 14, 818–829 (2008).

    CAS  PubMed  Google Scholar 

  27. Hu, G. et al. MTDH activation by 8q22 genomic gain promotes chemoresistance and metastasis of poor-prognosis breast cancer. Cancer Cell 15, 9–20 (2009). A study that mechanistically links the pro-metastatic gene metadherin with resistance to chemotherapy.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Stein, U. et al. MACC1, a newly identified key regulator of HGF–MET signaling, predicts colon cancer metastasis. Nature Med. 15, 59–67 (2009).

    CAS  PubMed  Google Scholar 

  29. Guo, C. et al. The noncoding RNA, miR-126, suppresses the growth of neoplastic cells by targeting phosphatidylinositol 3-kinase signaling and is frequently lost in colon cancers. Genes Chromosomes Cancer 47, 939–946 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Mundy, G. R. Metastasis to bone: causes, consequences and therapeutic opportunities. Nature Rev. Cancer 2, 584–593 (2002).

    CAS  Google Scholar 

  32. Lee, Y. T. Patterns of metastasis and natural courses of breast carcinoma. Cancer Metastasis Rev. 4, 153–172 (1985).

    CAS  PubMed  Google Scholar 

  33. Johansson, J. E. et al. Natural history of early, localized prostate cancer. JAMA 291, 2713–2719 (2004).

    CAS  PubMed  Google Scholar 

  34. Nieto, J., Grossbard, M. L. & Kozuch, P. Metastatic pancreatic cancer 2008: is the glass less empty? Oncologist 13, 562–576 (2008).

    CAS  PubMed  Google Scholar 

  35. Fearon, E. R. & Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 61, 759–767 (1990).

    CAS  PubMed  Google Scholar 

  36. Kinzler, K. W. & Vogelstein, B. Lessons from hereditary colorectal cancer. Cell 87, 159–170 (1996).

    CAS  PubMed  Google Scholar 

  37. Vogelstein, B. et al. Genetic alterations during colorectal-tumor development. N. Engl. J. Med. 319, 525–532 (1988).

    CAS  PubMed  Google Scholar 

  38. Samuels, Y. et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 304, 554 (2004).

    CAS  PubMed  Google Scholar 

  39. Baker, S. J. et al. Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas. Science 244, 217–221 (1989).

    CAS  PubMed  Google Scholar 

  40. Markowitz, S. et al. Inactivation of the type II TGF-β receptor in colon cancer cells with microsatellite instability. Science 268, 1336–1338 (1995).

    CAS  PubMed  Google Scholar 

  41. Jones, S. et al. Comparative lesion sequencing provides insights into tumor evolution. Proc. Natl Acad. Sci. USA 105, 4283–4288 (2008). A study that evaluated the frequency and timing of somatic mutations to estimate the clinical course of colorectal metastatic progression.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kedrin, D., van Rheenen, J., Hernandez, L., Condeelis, J. & Segall, J. E. Cell motility and cytoskeletal regulation in invasion and metastasis. J. Mammary Gland Biol. Neoplasia 12, 143–152 (2007).

    PubMed  Google Scholar 

  43. Weber, G. F. Molecular mechanisms of metastasis. Cancer Lett. 270, 181–190 (2008).

    CAS  PubMed  Google Scholar 

  44. Clark, E. A., Golub, T. R., Lander, E. S. & Hynes, R. O. Genomic analysis of metastasis reveals an essential role for RhoC. Nature 406, 532–535 (2000).

    CAS  PubMed  Google Scholar 

  45. Ewald, A. J., Brenot, A., Duong, M., Chan, B. S. & Werb, Z. Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis. Dev. Cell 14, 570–581 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Kouros-Mehr, H. et al. GATA-3 links tumor differentiation and dissemination in a luminal breast cancer model. Cancer Cell 13, 141–152 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Podsypanina, K. et al. Seeding and propagation of untransformed mouse mammary cells in the lung. Science 321, 1841–1844 (2008). This paper showed that phenotypically normal mouse mammary cells introduced into the mouse circulation can infiltrate the lungs and survive, leading to tumour initiation.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Ince, T. A. et al. Transformation of different human breast epithelial cell types leads to distinct tumor phenotypes. Cancer Cell 12, 160–170 (2007). This study showed that there are intrinsic differences in the tumorigenic and metastatic capabilities of different mammary cell types.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Bandyopadhyay, S. et al. Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression. Nature Med. 12, 933–938 (2006).

    CAS  PubMed  Google Scholar 

  51. Karpatkin, S. & Pearlstein, E. Role of platelets in tumor cell metastases. Ann. Intern. Med. 95, 636–641 (1981).

    CAS  PubMed  Google Scholar 

  52. Nieswandt, B., Hafner, M., Echtenacher, B. & Mannel, D. N. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 59, 1295–1300 (1999).

    CAS  PubMed  Google Scholar 

  53. Im, J. H. et al. Coagulation facilitates tumor cell spreading in the pulmonary vasculature during early metastatic colony formation. Cancer Res. 64, 8613–8619 (2004).

    CAS  PubMed  Google Scholar 

  54. Jain, S. et al. Platelet glycoprotein Ibα supports experimental lung metastasis. Proc. Natl Acad. Sci. USA 104, 9024–9028 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Paku, S., Dome, B., Toth, R. & Timar, J. Organ-specificity of the extravasation process: an ultrastructural study. Clin. Exp. Metastasis 18, 481–492 (2000).

    CAS  PubMed  Google Scholar 

  56. Lalor, P. F., Lai, W. K., Curbishley, S. M., Shetty, S. & Adams, D. H. Human hepatic sinusoidal endothelial cells can be distinguished by expression of phenotypic markers related to their specialised functions in vivo. World J. Gastroenterol. 12, 5429–5439 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Schluter, K. et al. Organ-specific metastatic tumor cell adhesion and extravasation of colon carcinoma cells with different metastatic potential. Am. J. Pathol. 169, 1064–1073 (2006).

    PubMed  PubMed Central  Google Scholar 

  58. Brown, D. M. & Ruoslahti, E. Metadherin, a cell surface protein in breast tumors that mediates lung metastasis. Cancer Cell 5, 365–374 (2004). Using phage-display libraries — a technology that had previously allowed this group to identify tissue-specific vasculature differences (or 'zipcodes') — this paper identifies metadherin as a lung-specific homing molecule.

    CAS  PubMed  Google Scholar 

  59. Kopp, H. G., Avecilla, S. T., Hooper, A. T. & Rafii, S. The bone marrow vascular niche: home of HSC differentiation and mobilization. Physiology 20, 349–356 (2005).

    CAS  PubMed  Google Scholar 

  60. Weis, S., Cui, J., Barnes, L. & Cheresh, D. Endothelial barrier disruption by VEGF-mediated Src activity potentiates tumor cell extravasation and metastasis. J. Cell Biol. 167, 223–229 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Gupta, G. P. et al. Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature 446, 765–770 (2007).

    CAS  PubMed  Google Scholar 

  62. Karnoub, A. E. et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449, 557–563 (2007).

    CAS  PubMed  Google Scholar 

  63. Padua, D. et al. TGFβ primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell 133, 66–77 (2008). References 62 and 63 exemplify how paracrine signals from the stroma of a primary tumour can stimulate departing cancer cells to extravasate into the lung without affecting primary tumorigenesis.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang, H. et al. Tumor cell α3β1 integrin and vascular laminin-5 mediate pulmonary arrest and metastasis. J. Cell Biol. 164, 935–941 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Weil, R. J., Palmieri, D. C., Bronder, J. L., Stark, A. M. & Steeg, P. S. Breast cancer metastasis to the central nervous system. Am. J. Pathol. 167, 913–920 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Minn, A. J. et al. Lung metastasis genes couple breast tumor size and metastatic spread. Proc. Natl Acad. Sci. USA 104, 6740–6745 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Smid, M. et al. Genes associated with breast cancer metastatic to bone. J. Clin. Oncol. 24, 2261–2267 (2006).

    CAS  PubMed  Google Scholar 

  68. Nesbitt, J. C., Putnam, J. B. Jr, Walsh, G. L., Roth, J. A. & Mountain, C. F. Survival in early-stage non-small cell lung cancer. Ann. Thorac. Surg. 60, 466–472 (1995).

    CAS  PubMed  Google Scholar 

  69. Ries, L. A. G. et al. SEER Cancer Statistics Review, 1975–2005 National Cancer Institute [online] http://seer.cancer.gov/csr/1975_2005/index.html (2008)

    Google Scholar 

  70. Janne, P. A. et al. Twenty-five years of clinical research for patients with limited-stage small cell lung carcinoma in North America. Cancer 95, 1528–1538 (2002).

    PubMed  Google Scholar 

  71. Briele, H. A. & Das Gupta, T. K. Natural history of cutaneous malignant melanoma. World J. Surg. 3, 255–270 (1979).

    CAS  PubMed  Google Scholar 

  72. Sorlie, T. et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl Acad. Sci. USA 98, 10869–10874 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Garraway, L. A. et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436, 117–122 (2005).

    CAS  PubMed  Google Scholar 

  74. Gupta, P. B. et al. The melanocyte differentiation program predisposes to metastasis after neoplastic transformation. Nature Genet. 37, 1047–1054 (2005).

    CAS  PubMed  Google Scholar 

  75. Wong, D. J. et al. Module map of stem cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell 2, 333–344 (2008). This study shows an association between the expression of embryonic stem cell gene modules in primary tumours and increased metastatic potential.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Varambally, S. et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419, 624–629 (2002).

    CAS  PubMed  Google Scholar 

  77. Kleer, C. G. et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc. Natl Acad. Sci. USA 100, 11606–11611 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Varambally, S. et al. Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science 322, 1695–1699 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Gregory, P. A. et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nature Cell Biol. 10, 593–601 (2008).

    CAS  PubMed  Google Scholar 

  80. Ma, L., Teruya-Feldstein, J. & Weinberg, R. A. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 449, 682–688 (2007).

    CAS  PubMed  Google Scholar 

  81. Demicheli, R. Tumour dormancy: findings and hypotheses from clinical research on breast cancer. Semin. Cancer Biol. 11, 297–306 (2001).

    CAS  PubMed  Google Scholar 

  82. Braun, S. et al. Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N. Engl. J. Med. 342, 525–533 (2000).

    CAS  PubMed  Google Scholar 

  83. Husemann, Y. et al. Systemic spread is an early step in breast cancer. Cancer Cell 13, 58–68 (2008). This paper shows that dissemination of tumour cells can occur at early stages of primary tumour development in ERBB2 and PyMT mouse models. Moreover, transplantation of pre-malignant DTCs into recipient bone marrow releases these cells from dormancy.

    PubMed  Google Scholar 

  84. White, D. E. et al. Targeted disruption of β1-integrin in a transgenic mouse model of human breast cancer reveals an essential role in mammary tumor induction. Cancer Cell 6, 159–170 (2004).

    CAS  PubMed  Google Scholar 

  85. Aguirre-Ghiso, J. A., Estrada, Y., Liu, D. & Ossowski, L. ERKMAPK activity as a determinant of tumor growth and dormancy; regulation by p38SAPK. Cancer Res. 63, 1684–1695 (2003).

    CAS  PubMed  Google Scholar 

  86. Aguirre-Ghiso, J. A., Ossowski, L. & Rosenbaum, S. K. Green fluorescent protein tagging of extracellular signal-regulated kinase and p38 pathways reveals novel dynamics of pathway activation during primary and metastatic growth. Cancer Res. 64, 7336–7345 (2004).

    CAS  PubMed  Google Scholar 

  87. Nash, K. T. et al. Requirement of KISS1 secretion for multiple organ metastasis suppression and maintenance of tumor dormancy. J. Natl Cancer Inst. 99, 309–321 (2007).

    CAS  PubMed  Google Scholar 

  88. Xu, L., Begum, S., Hearn, J. D. & Hynes, R. O. GPR56, an atypical G protein-coupled receptor, binds tissue transglutaminase, TG2, and inhibits melanoma tumor growth and metastasis. Proc. Natl Acad. Sci. USA 103, 9023–9028 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Park, Y. G. et al. Sipa1 is a candidate for underlying the metastasis efficiency modifier locus Mtes1. Nature Genet. 37, 1055–1062 (2005).

    CAS  PubMed  Google Scholar 

  90. Holmgren, L., O'Reilly, M. S. & Folkman, J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nature Med. 1, 149–153 (1995).

    CAS  PubMed  Google Scholar 

  91. Almog, N. et al. Transcriptional switch of dormant tumors to fast-growing angiogenic phenotype. Cancer Res. 69, 836–844 (2009).

    CAS  PubMed  Google Scholar 

  92. Naumov, G. N., Akslen, L. A. & Folkman, J. Role of angiogenesis in human tumor dormancy: animal models of the angiogenic switch. Cell Cycle 5, 1779–1787 (2006).

    CAS  PubMed  Google Scholar 

  93. Luzzi, K. J. et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am. J. Pathol. 153, 865–873 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Becker, S., Becker-Pergola, G., Wallwiener, D., Solomayer, E. F. & Fehm, T. Detection of cytokeratin-positive cells in the bone marrow of breast cancer patients undergoing adjuvant therapy. Breast Cancer Res. Treat. 97, 91–96 (2006).

    CAS  PubMed  Google Scholar 

  95. Ling, L. J. et al. A novel mouse model of human breast cancer stem-like cells with high CD44+CD24−/lower phenotype metastasis to human bone. Chin. Med. J. 121, 1980–1986 (2008).

    CAS  PubMed  Google Scholar 

  96. Gupta, G. P. et al. ID genes mediate tumor reinitiation during breast cancer lung metastasis. Proc. Natl Acad. Sci. USA 104, 19506–19511 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Swarbrick, A., Roy, E., Allen, T. & Bishop, J. M. Id1 cooperates with oncogenic Ras to induce metastatic mammary carcinoma by subversion of the cellular senescence response. Proc. Natl Acad. Sci. USA 105, 5402–5407 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Park, B. K. et al. NF-κB in breast cancer cells promotes osteolytic bone metastasis by inducing osteoclastogenesis via GM-CSF. Nature Med. 13, 62–69 (2007).

    CAS  PubMed  Google Scholar 

  99. Fitzgerald, D. P. et al. Reactive glia are recruited by highly proliferative brain metastases of breast cancer and promote tumor cell colonization. Clin. Exp. Metastasis 25, 799–810 (2008).

    PubMed  PubMed Central  Google Scholar 

  100. Sharma, S. V., Bell, D. W., Settleman, J. & Haber, D. A. Epidermal growth factor receptor mutations in lung cancer. Nature Rev. Cancer 7, 169–181 (2007).

    CAS  Google Scholar 

  101. Engelman, J. A. et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316, 1039–1043 (2007).

    CAS  PubMed  Google Scholar 

  102. Bean, J. et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc. Natl Acad. Sci. USA 104, 20932–20937 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Aragon-Ching, J. B. & Zujewski, J. A. CNS metastasis: an old problem in a new guise. Clin. Cancer Res. 13, 1644–1647 (2007).

    CAS  PubMed  Google Scholar 

  104. Fidler, I. J., Yano, S., Zhang, R. D., Fujimaki, T. & Bucana, C. D. The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol. 3, 53–57 (2002).

    CAS  PubMed  Google Scholar 

  105. Wiedswang, G. et al. Isolated tumor cells in bone marrow three years after diagnosis in disease-free breast cancer patients predict unfavorable clinical outcome. Clin. Cancer Res. 10, 5342–5348 (2004).

    PubMed  Google Scholar 

  106. Erler, J. T. et al. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 440, 1222–1226 (2006).

    CAS  PubMed  Google Scholar 

  107. 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). References 106 and 107 describe the metastasis progression gene LOX and how it can be induced in the primary tumour to mediate one function at the primary site and a different function in secondary organs, both of which are required for metastasis.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  110. Condeelis, J. & Pollard, J. W. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 124, 263–266 (2006).

    CAS  PubMed  Google Scholar 

  111. Kim, S. et al. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 457, 102–106 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Bierie, B. & Moses, H. L. Tumour microenvironment: TGFβ: the molecular Jekyll and Hyde of cancer. Nature Rev. Cancer 6, 506–520 (2006).

    CAS  PubMed  Google Scholar 

  113. Braun, S. et al. A pooled analysis of bone marrow micrometastasis in breast cancer. N. Engl. J. Med. 353, 793–802 (2005).

    CAS  PubMed  Google Scholar 

  114. Wikman, H., Vessella, R. & Pantel, K. Cancer micrometastasis and tumour dormancy. APMIS 116, 754–770 (2008).

    CAS  PubMed  Google Scholar 

  115. Pantel, K. & Brakenhoff, R. H. Dissecting the metastatic cascade. Nature Rev. Cancer 4, 448–456 (2004).

    CAS  Google Scholar 

  116. Klein, C. A. et al. Genetic heterogeneity of single disseminated tumour cells in minimal residual cancer. Lancet 360, 683–689 (2002).

    CAS  PubMed  Google Scholar 

  117. Schardt, J. A. et al. Genomic analysis of single cytokeratin-positive cells from bone marrow reveals early mutational events in breast cancer. Cancer Cell 8, 227–239 (2005).

    CAS  PubMed  Google Scholar 

  118. Ramaswamy, S., Ross, K. N., Lander, E. S. & Golub, T. R. A molecular signature of metastasis in primary solid tumors. Nature Genet. 33, 49–54 (2003).

    CAS  PubMed  Google Scholar 

  119. Weigelt, B. et al. Gene expression profiles of primary breast tumors maintained in distant metastases. Proc. Natl Acad. Sci. USA 100, 15901–15905 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Massagué lab for insightful discussions. The primary research for the topic of this review is supported by grants from the National Institutes of Health, the Hearst Foundation and the Kleberg Foundation. D.X.N was a postdoctoral fellow of the Damon Runyon Cancer Research Foundation. J.M. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joan Massagué.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Joan massagué's homepage

Glossary

Infiltration

The entry of cancer cells into distant organs through invasion and extravasation.

Colonization

The outgrowth of metastatic cells that have co-opted a distant organ microenvironment.

Latency

The time between primary tumour diagnosis and clinically detectable metastatic outgrowths.

Intravasation

The entry of tumour cells into the bloodstream.

Extravasation

The exit of tumour cells from capillary beds into the parenchyma of an organ.

Basal breast cancer

A more aggressive subtype of breast cancer with characteristics of mammary basal cells, and that typically lacks oestrogen and progesterone receptors.

Luminal breast cancer

A subtype of breast cancer with characteristics of cells that originate from the normal lumen or ducts of the mammary gland.

Dormancy

A state of cellular quiescence in the G0 phase of the cell cycle. When referring to a tumour cell mass, dormancy describes a balanced state of proliferation and apoptosis.

Angiogenic switch

The transition between a non-angiogenic state of the tumour cell mass and a neovascularized state that enables tumour oxygenation and growth.

Tumour-propagating phenotype

The ability of the infiltrated tumour cells to reinitiate growth at the secondary site. This is referred to by some investigators as the 'cancer stem cell phenotype'.

Metastatic speciation

An evolutionary process by which new metastatic populations arise, owing to the various selective pressures that act on the heterogeneous cancer cells escaping the primary tumour.

Gliosis

Stimulation of astrocytes in injured areas of the brain.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nguyen, D., Bos, P. & Massagué, J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9, 274–284 (2009). https://doi.org/10.1038/nrc2622

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc2622

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing