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: a question of life or death

Key Points

  • Metastasis occurs through a series of sequential steps in which tumour cells first migrate from the primary tumour, penetrate blood vessels and then colonize distant sites. It is a highly inefficient process. Indeed, very few of the tumour cells that gain access to the vasculature give rise to metastastic foci in a secondary organ.

  • Recent data indicate that the mechanisms controlling metastasis can be regulated independently from primary tumour development.

  • In vitro and in vivo, the metastatic potential of tumours is associated with an increased resistance to apoptosis. Furthermore, the experimental modulation of apoptotic or anti-apoptotic factors influences metastatic efficiency.

  • Anoikis and amorphosis are important barriers to metastasis. Anoikis is cell death induced by the disruption of cell attachment and cell–matrix interactions, whereas amorphosis is cell death stimulated by the loss of cytoskeletal architecture.

  • Early survival of tumour cells after attachment to the secondary site and the development of micrometastases are crucial steps of the metastatic process.

  • Metastasis is the most common cause of cancer death. Most patients with metastatic disease respond transiently to conventional treatments. Further elucidation of the relationship between resistance to apoptosis of metastatic cancer cells and their chemoresistance should provide important clues to improve systemic therapies.

Abstract

The metastatic process is highly inefficient — very few of the many cells that migrate from the primary tumour successfully colonize distant sites. One proposed mechanism to explain this inefficiency is provided by the cancer stem cell model, which hypothesizes that micrometastases can only be established by tumour stem cells, which are few in number. However, recent in vitro and in vivo observations indicate that apoptosis is an important process regulating metastasis. Here we stress that the inhibition of cell death, apart from its extensively described function in primary tumour development, is a crucial characteristic of metastatic cancer 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: Apoptosis as a safeguard system preventing metastasis at three crucial steps.
Figure 2: Resistance to anoikis and amorphosis is a crucial event during metastatic spread.
Figure 3: Tumour immune surveillance.
Figure 4: Role of the microenvironment in the development of micrometastases.
Figure 5: The dependence receptor model and metastasis.

Similar content being viewed by others

References

  1. Weigelt, B., Peterse, J. L. & van 't Veer, L. J. Breast cancer metastasis: markers and models. Nature Rev. Cancer 5, 591–602 (2005).

    CAS  Google Scholar 

  2. Salvesen, G. S. & Dixit, V. M. Caspases: intracellular signaling by proteolysis. Cell 91, 443–446 (1997).

    CAS  PubMed  Google Scholar 

  3. Zimmermann, K. C. & Green, D. R. How cells die: apoptosis pathways. J. Allergy Clin. Immunol. 108, S99–S103 (2001).

    CAS  PubMed  Google Scholar 

  4. Salvesen, G. S. & Duckett, C. S. IAP proteins: blocking the road to death's door. Nature Rev. Mol. Cell Biol. 3, 401–410 (2002).

    CAS  Google Scholar 

  5. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000). A seminal review describing the main rules that govern the transformation of normal human cells into malignant cancers.

    CAS  PubMed  Google Scholar 

  6. Yin, C., Knudson, C. M., Korsmeyer, S. J. & Van Dyke, T. Bax suppresses tumorigenesis and stimulates apoptosis in vivo. Nature 385, 637–640 (1997).

    CAS  PubMed  Google Scholar 

  7. Lowe, S. W., Cepero, E. & Evan, G. Intrinsic tumour suppression. Nature 432, 307–315 (2004).

    CAS  PubMed  Google Scholar 

  8. Fidler, I. J. & Nicolson, G. L. Fate of recirculating B16 melanoma metastatic variant cells in parabiotic syngeneic recipients. J. Natl Cancer Inst. 58, 1867–1872 (1977).

    CAS  PubMed  Google Scholar 

  9. Liotta, L. A., Vembu, D., Saini, R. K. & Boone, C. In vivo monitoring of the death rate of artificial murine pulmonary micrometastases. Cancer Res. 38, 1231–1236 (1978).

    CAS  PubMed  Google Scholar 

  10. Varani, J., Lovett, E. J., Elgebaly, S., Lundy, J. & Ward, P. A. In vitro and in vivo adherence of tumor cell variants correlated with tumor formation. Am. J. Pathol. 101, 345–352 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Inbal, B. et al. DAP kinase links the control of apoptosis to metastasis. Nature 390, 180–184 (1997). By using lung carcinoma clones, this study shows that the inhibition of the expression of DAPK, a positive mediator of apoptosis, favours the metastatic process.

    CAS  PubMed  Google Scholar 

  12. Um, J. H. et al. Relationship between antiapoptotic molecules and metastatic potency and the involvement of DNA-dependent protein kinase in the chemosensitization of metastatic human cancer cells by epidermal growth factor receptor blockade. J. Pharmacol. Exp. Ther. 311, 1062–1070 (2004).

    CAS  PubMed  Google Scholar 

  13. Glinsky, G. V., Glinsky, V. V., Ivanova, A. B. & Hueser, C. J. Apoptosis and metastasis: increased apoptosis resistance of metastatic cancer cells is associated with the profound deficiency of apoptosis execution mechanisms. Cancer Lett. 115, 185–193 (1997). Data showing that the metastatic potential of murine and human cancer cells is strictly associated with the increased resistance to apoptosis.

    CAS  PubMed  Google Scholar 

  14. Del Bufalo, D., Biroccio, A., Leonetti, C. & Zupi, G. Bcl-2 overexpression enhances the metastatic potential of a human breast cancer line. FASEB J. 11, 947–953 (1997). An elegant study showing that the overexpression of the anti-apoptotic oncoprotein BCL2 increases the metastatic potential of human breast cancer cells.

    CAS  PubMed  Google Scholar 

  15. McConkey, D. J., Greene, G. & Pettaway, C. A. Apoptosis resistance increases with metastatic potential in cells of the human LNCaP prostate carcinoma line. Cancer Res. 56, 5594–5599 (1996).

    CAS  PubMed  Google Scholar 

  16. Furuya, Y., Krajewski, S., Epstein, J. I., Reed, J. C. & Isaacs, J. T. Expression of bcl-2 and the progression of human and rodent prostatic cancers. Clin. Cancer Res. 2, 389–398 (1996).

    CAS  PubMed  Google Scholar 

  17. Owen-Schaub, L. B., van Golen, K. L., Hill, L. L. & Price, J. E. Fas and Fas ligand interactions suppress melanoma lung metastasis. J. Exp. Med. 188, 1717–1723 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Thompson, T. C. et al. Loss of p53 function leads to metastasis in ras+myc-initiated mouse prostate cancer. Oncogene 10, 869–879 (1995).

    CAS  PubMed  Google Scholar 

  19. Silvestrini, R. et al. Validation of p53 accumulation as a predictor of distant metastasis at 10 years of follow-up in 1400 node-negative breast cancers. Clin. Cancer Res. 2, 2007–2013 (1996).

    CAS  PubMed  Google Scholar 

  20. Lewis, B. C. et al. The absence of p53 promotes metastasis in a novel somatic mouse model for hepatocellular carcinoma. Mol. Cell. Biol. 25, 1228–1237 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Ko, J. et al. Transgenic mouse model for breast cancer: induction of breast cancer in novel oncogene HCCR-2 transgenic mice. Oncogene 23, 1950–1953 (2004).

    CAS  PubMed  Google Scholar 

  22. Esteller, M. Relevance of DNA methylation in the management of cancer. Lancet Oncol. 4, 351–358 (2003).

    CAS  PubMed  Google Scholar 

  23. Bao, S. et al. Periostin potently promotes metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. Cancer Cell 5, 329–339 (2004).

    CAS  PubMed  Google Scholar 

  24. Martin, S. S. & Vuori, K. Regulation of Bcl-2 proteins during anoikis and amorphosis. Biochim. Biophys. Acta 1692, 145–157 (2004). A detailed analysis of the molecular mechanisms of apoptosis induced during anoikis and amorphosis.

    CAS  PubMed  Google Scholar 

  25. Streuli, C. H. & Gilmore, A. P. Adhesion-mediated signaling in the regulation of mammary epithelial cell survival. J. Mammary Gland. Biol. Neoplasia 4, 183–191 (1999).

    CAS  PubMed  Google Scholar 

  26. Martin, S. S. & Leder, P. Human MCF10A mammary epithelial cells undergo apoptosis following actin depolymerization that is independent of attachment and rescued by Bcl-2. Mol. Cell. Biol. 21, 6529–6536 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Puthalakath, H., Huang, D. C., O'Reilly, L. A., King, S. M. & Strasser, A. The proapoptotic activity of the Bcl-2 family member Bim is regulated by interaction with the dynein motor complex. Mol. Cell 3, 287–296 (1999).

    CAS  PubMed  Google Scholar 

  28. Puthalakath, H. et al. Bmf: a proapoptotic BH3-only protein regulated by interaction with the myosin V actin motor complex, activated by anoikis. Science 293, 1829–1832 (2001).

    CAS  PubMed  Google Scholar 

  29. Zhu, Z. et al. Anoikis and metastatic potential of cloudman S91 melanoma cells. Cancer Res. 61, 1707–1716 (2001).

    CAS  PubMed  Google Scholar 

  30. Owens, L. V. et al. Overexpression of the focal adhesion kinase (p125FAK) in invasive human tumors. Cancer Res. 55, 2752–2755 (1995).

    CAS  PubMed  Google Scholar 

  31. McLean, G. W. et al. Specific deletion of focal adhesion kinase suppresses tumor formation and blocks malignant progression. Genes Dev. 18, 2998–3003 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Gentile, A. & Comoglio, P. M. Invasive growth: a genetic program. Int. J. Dev. Biol. 48, 451–456 (2004).

    CAS  PubMed  Google Scholar 

  33. Camp, E. R. et al. RON, a tyrosine kinase receptor involved in tumor progression and metastasis. Ann. Surg. Oncol. 12, 273–281 (2005).

    PubMed  Google Scholar 

  34. Peace, B. E., Toney-Earley, K., Collins, M. H. & Waltz, S. E. Ron receptor signaling augments mammary tumor formation and metastasis in a murine model of breast cancer. Cancer Res. 65, 1285–1293 (2005).

    CAS  PubMed  Google Scholar 

  35. Zeng, Q. et al. Hepatocyte growth factor inhibits anoikis in head and neck squamous cell carcinoma cells by activation of ERK and Akt signaling independent of NFκB. J. Biol. Chem. 277, 25203–25208 (2002).

    CAS  PubMed  Google Scholar 

  36. Pinkas, J., Martin, S. S. & Leder, P. Bcl-2-mediated cell survival promotes metastasis of EpH4 βMEKDD mammary epithelial cells. Mol. Cancer Res. 2, 551–556 (2004).

    CAS  PubMed  Google Scholar 

  37. Martin, S. S. et al. A cytoskeleton-based functional genetic screen identifies Bcl-xL as an enhancer of metastasis, but not primary tumor growth. Oncogene 23, 4641–4645 (2004). An elegant study identifying BCL-X L as a suppressor of cytoskeleton-dependent death and that overexpression in mouse mammary epithelial cells does not induce primary tumour formation but strongly increases metastatic potential.

    CAS  PubMed  Google Scholar 

  38. Olopade, O. I. et al. Overexpression of BCL-x protein in primary breast cancer is associated with high tumor grade and nodal metastases. Cancer J. Sci. Am. 3, 230–237 (1997).

    CAS  PubMed  Google Scholar 

  39. Fernandez, Y., Gu, B., Martinez, A., Torregrosa, A. & Sierra, A. Inhibition of apoptosis in human breast cancer cells: role in tumor progression to the metastatic state. Int. J. Cancer 101, 317–326 (2002).

    CAS  PubMed  Google Scholar 

  40. Berezovskaya, O. et al. Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. Cancer Res. 65, 2378–2386 (2005). Comprehensive work showing that the apoptosis-inhibitory protein XIAP contributes to the anoikis resistance of circulating metastatic prostate cancer cells.

    CAS  PubMed  Google Scholar 

  41. Shen, T. L. et al. Conditional knockout of focal adhesion kinase in endothelial cells reveals its role in angiogenesis and vascular development in late embryogenesis. J. Cell Biol. 169, 941–952 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Wyckoff, J. B., Jones, J. G., Condeelis, J. S. & Segall, J. E. A critical step in metastasis: in vivo analysis of intravasation at the primary tumor. Cancer Res. 60, 2504–2511 (2000).

    CAS  PubMed  Google Scholar 

  43. Fidler, I. J. Metastasis: guantitative analysis of distribution and fate of tumor embolilabeled with 125 I-5-iodo-2′-deoxyuridine. J. Natl Cancer Inst. 45, 773–782 (1970).

    CAS  PubMed  Google Scholar 

  44. Wong, C. W. et al. Apoptosis: an early event in metastatic inefficiency. Cancer Res. 61, 333–338 (2001).

    CAS  PubMed  Google Scholar 

  45. 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). References 43, 44 and 45 examine the fate of cancer cells in the blood stream and in the site of metastasis, aiming to identify mechanisms explaining the overall metastatic inefficiency.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Jakobisiak, M., Lasek, W. & Golab, J. Natural mechanisms protecting against cancer. Immunol. Lett. 90, 103–122 (2003).

    CAS  PubMed  Google Scholar 

  47. Pardoll, D. Does the immune system see tumors as foreign or self? Annu. Rev. Immunol. 21, 807–839 (2003).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  49. Rosenberg, S. A. et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N. Engl. J. Med. 313, 1485–1492 (1985).

    CAS  PubMed  Google Scholar 

  50. Rosenberg, S. A. Progress in human tumour immunology and immunotherapy. Nature 411, 380–384 (2001).

    CAS  PubMed  Google Scholar 

  51. Kim, S., Iizuka, K., Aguila, H. L., Weissman, I. L. & Yokoyama, W. M. In vivo natural killer cell activities revealed by natural killer cell-deficient mice. Proc. Natl Acad. Sci. USA 97, 2731–2736 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Smyth, M. J. et al. NKG2D recognition and perforin effector function mediate effective cytokine immunotherapy of cancer. J. Exp. Med. 200, 1325–1335 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Takeda, K. et al. Involvement of tumor necrosis factor-related apoptosis-inducing ligand in NK cell-mediated and IFN-γ-dependent suppression of subcutaneous tumor growth. Cell Immunol. 214, 194–200 (2001).

    CAS  PubMed  Google Scholar 

  54. Cretney, E. et al. Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice. J. Immunol. 168, 1356–1361 (2002).

    CAS  PubMed  Google Scholar 

  55. Weiss, L., Elkin, G. & Barbera-Guillem, E. The differential resistance of B16 wild-type and F10 cells to mechanical trauma in vitro. Invasion Metastasis 13, 92–101 (1993).

    CAS  PubMed  Google Scholar 

  56. Ziegler, T., Silacci, P., Harrison, V. J. & Hayoz, D. Nitric oxide synthase expression in endothelial cells exposed to mechanical forces. Hypertension 32, 351–355 (1998).

    CAS  PubMed  Google Scholar 

  57. Wang, H. H. et al. B16 melanoma cell arrest in the mouse liver induces nitric oxide release and sinusoidal cytotoxicity: a natural hepatic defense against metastasis. Cancer Res. 60, 5862–5869 (2000).

    CAS  PubMed  Google Scholar 

  58. Edmiston, K. H. et al. Role of nitric oxide and superoxide anion in elimination of low metastatic human colorectal carcinomas by unstimulated hepatic sinusoidal endothelial cells. Cancer Res. 58, 1524–1531 (1998).

    CAS  PubMed  Google Scholar 

  59. Jaattela, M., Wissing, D., Kokholm, K., Kallunki, T. & Egeblad, M. Hsp70 exerts its anti-apoptotic function downstream of caspase-3-like proteases. EMBO J. 17, 6124–6134 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Kluger, H. M. et al. Using a xenograft model of human breast cancer metastasis to find genes associated with clinically aggressive disease. Cancer Res. 65, 5578–5587 (2005).

    CAS  PubMed  Google Scholar 

  61. Schmitt, C. A. et al. Dissecting p53 tumor suppressor functions in vivo. Cancer Cell 1, 289–298 (2002).

    CAS  PubMed  Google Scholar 

  62. Nikiforov, M. A. et al. p53 modulation of anchorage independent growth and experimental metastasis. Oncogene 13, 1709–1719 (1996). Data showing that the inactivation of p53 in transformed mouse embryonic fibroblasts facilitates experimental metastasis by promoting the survival of tumour cells in circulation.

    CAS  PubMed  Google Scholar 

  63. Schuler, M. & Green, D. R. Transcription, apoptosis and p53: catch-22. Trends Genet. 21, 182–187 (2005).

    CAS  PubMed  Google Scholar 

  64. Ponta, H., Sherman, L. & Herrlich, P. A. CD44: from adhesion molecules to signalling regulators. Nature Rev. Mol. Cell Biol. 4, 33–45 (2003).

    CAS  Google Scholar 

  65. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. O'Reilly, M. S. et al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 79, 315–328 (1994).

    CAS  PubMed  Google Scholar 

  67. O'Reilly, M. S., Holmgren, L., Chen, C. & Folkman, J. Angiostatin induces and sustains dormancy of human primary tumors in mice. Nature Med. 2, 689–692 (1996).

    CAS  PubMed  Google Scholar 

  68. O'Reilly, M. S. et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 88, 277–285 (1997).

    CAS  PubMed  Google Scholar 

  69. Giehl, K. Oncogenic Ras in tumour progression and metastasis. Biol. Chem. 386, 193–205 (2005).

    CAS  PubMed  Google Scholar 

  70. Liao, Y. et al. Modulation of apoptosis, tumorigenesity and metastatic potential with antisense H-ras oligodeoxynucleotides in a high metastatic tumor model of hepatoma: LCI-D20. Hepatogastroenterology 47, 365–370 (2000).

    CAS  PubMed  Google Scholar 

  71. Varghese, H. J. et al. Activated ras regulates the proliferation/apoptosis balance and early survival of developing micrometastases. Cancer Res. 62, 887–891 (2002). By using in vivo video microscopy and detailed cell quantification, this study provides evidence for a direct role for Ras in the maintenance of metastatic growth by mediating a shift in the proliferation/apoptosis balance.

    CAS  PubMed  Google Scholar 

  72. Mehlen, P. The dependence receptor notion: another way to see death. Cell Death Differ. 12, 1003 (2005).

    CAS  PubMed  Google Scholar 

  73. Mehlen, P. & Bredesen, D. E. The dependence receptor hypothesis. Apoptosis 9, 37–49 (2004).

    CAS  PubMed  Google Scholar 

  74. Mehlen, P. & Thibert, C. Dependence receptors: between life and death. Cell Mol. Life Sci. 61, 1854–1866 (2004).

    CAS  PubMed  Google Scholar 

  75. Bredesen, D. E., Mehlen, P. & Rabizadeh, S. Receptors that mediate cellular dependence. Cell Death Differ. 12, 1031–1043 (2005).

    CAS  PubMed  Google Scholar 

  76. Wang, J. J. et al. Dimerization-dependent block of the proapoptotic effect of p75(NTR). J. Neurosci. Res. 60, 587–593 (2000).

    CAS  PubMed  Google Scholar 

  77. Forcet, C. et al. The dependence receptor DCC (deleted in colorectal cancer) defines an alternative mechanism for caspase activation. Proc. Natl Acad. Sci. USA 98, 3416–3421 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Stupack, D. G., Puente, X. S., Boutsaboualoy, S., Storgard, C. M. & Cheresh, D. A. Apoptosis of adherent cells by recruitment of caspase-8 to unligated integrins. J. Cell Biol. 155, 459–470 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Mehlen, P. & Furne, C. Netrin-1: when a neuronal guidance cue turns out to be a regulator of tumorigenesis. Cell Mol. Life Sci. 62, 2599–2616 (2005).

    CAS  PubMed  Google Scholar 

  80. Fearon, E. R. et al. Identification of a chromosome 18q gene that is altered in colorectal cancers. Science 247, 49–56 (1990).

    CAS  PubMed  Google Scholar 

  81. Mehlen, P. & Fearon, E. R. Role of the dependence receptor DCC in colorectal cancer pathogenesis. J. Clin. Oncol. 22, 3420–3428 (2004).

    CAS  PubMed  Google Scholar 

  82. Thiebault, K. et al. The netrin-1 receptors UNC5H are putative tumor suppressors controlling cell death commitment. Proc. Natl Acad. Sci. USA 100, 4173–4178 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Mehlen, P. et al. The DCC gene product induces apoptosis by a mechanism requiring receptor proteolysis. Nature 395, 801–804 (1998).

    CAS  PubMed  Google Scholar 

  84. Llambi, F., Causeret, F., Bloch-Gallego, E. & Mehlen, P. Netrin-1 acts as a survival factor via its receptors UNC5H and DCC. EMBO J. 20, 2715–2722 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Mazelin, L. et al. Netrin-1 controls colorectal tumorigenesis by regulating apoptosis. Nature 431, 80–84 (2004). This paper suggests for the first time, using a mouse model, that dependence receptors might regulate tumorigenesis.

    CAS  PubMed  Google Scholar 

  86. Llambi, F. et al. The dependence receptor UNC5H2 mediates apoptosis through DAP-kinase. EMBO J. 24, 1192–1201 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Tanikawa, C., Matsuda, K., Fukuda, S., Nakamura, Y. & Arakawa, H. p53RDL1 regulates p53-dependent apoptosis. Nature Cell Biol. 5, 216–223 (2003).

    CAS  PubMed  Google Scholar 

  88. Stupack, D. G. Integrins as a distinct subtype of dependence receptors. Cell Death Differ. 12, 1021–1030 (2005).

    CAS  PubMed  Google Scholar 

  89. Stupack, D. G. et al. Potentiation of neuroblastoma metastasis by loss of caspase 8. Nature 439 95–99 (2006). This paper represents the first demonstration of a role for dependence receptors in metastasis regulation.

    CAS  PubMed  Google Scholar 

  90. Hofmann, U. B., Houben, R., Brocker, E. B. & Becker, J. C. Role of matrix metalloproteinases in melanoma cell invasion. Biochimie 87, 307–314 (2005).

    CAS  PubMed  Google Scholar 

  91. Abraham, R. et al. Identification of MMP-15 as an anti-apoptotic factor in cancer cells. J. Biol. Chem. 280, 34123–34132 (2005).

    CAS  PubMed  Google Scholar 

  92. Strand, S. et al. Cleavage of CD95 by matrix metalloproteinase-7 induces apoptosis resistance in tumour cells. Oncogene 23, 3732–3736 (2004).

    CAS  PubMed  Google Scholar 

  93. Huber, M. A. et al. NF-κB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J. Clin. Invest. 114, 569–581 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Chen, Z. F. & Behringer, R. R. Twist is required in head mesenchyme for cranial neural tube morphogenesis. Genes Dev. 9, 686–699 (1995).

    CAS  PubMed  Google Scholar 

  95. Yang, J. et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117, 927–939 (2004).

    CAS  PubMed  Google Scholar 

  96. Maestro, R. et al. Twist is a potential oncogene that inhibits apoptosis. Genes Dev. 13, 2207–2217 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Valsesia-Wittmann, S. et al. Oncogenic cooperation between H-Twist and N-Myc overrides failsafe programs in cancer cells. Cancer Cell 6, 625–630 (2004). References 95, 96 and 97 show the oncogenic properties of H-Twist, highlighting its role as an anti-apoptotic factor (references 96 and 97), and establishing its implication in the metastatic process (reference 95).

    CAS  PubMed  Google Scholar 

  98. Puisieux, A., Valsesia-Wittmann, S. & Ansieau, S. A twist for survival and cancer progression. Br. J. Cancer 94, 13–17 (2006).

    CAS  PubMed  Google Scholar 

  99. Muller, W. J., Sinn, E., Pattengale, P. K., Wallace, R. & Leder, P. Single-step induction of mammary adenocarcinoma in transgenic mice bearing the activated c-neu oncogene. Cell 54, 105–115 (1988).

    CAS  PubMed  Google Scholar 

  100. Greenberg, N. M., et al. Prostate cancer in a transgenic mouse. Proc. Natl Acad. Sci. USA 92, 3439–3443 (1995)

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Sierra, A. Metastases and their microenvironments: linking pathogenesis and therapy. Drug Resist. Updat. 8, 247–257 (2005).

    CAS  PubMed  Google Scholar 

  102. Fesik, S. W. Promoting apoptosis as a strategy for cancer drug discovery. Nature Rev. Cancer 5, 876–885 (2005).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Bilak for text correction, C. Caux for critical reading of the manuscript and B. Bouchet for the development of figures. Work in Mehlen's and Puisieux's laboratories are supported by institutional funds from the Centre National de la Recherche Scientifique (P.M.), Agence Nationale de la Recherche (ANR) (P.M.), Institut national de la santé et de la recherche médicale (A.P.), and Institut National du Cancer (INCA) (P.M. and A.P.), and by grants from the Ligue Nationale Contre le Cancer (P.M. and A.P.), the Association pour la Recherche contre le Cancer (P.M. and A.P.), the Comité Départemental de l'Ain (A.P.) and the National Institute of Health (P.M.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrick Mehlen.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

National Cancer Institute

colon cancer

breast cancer

Glossary

Natural killer cells

Large granular lymphocytes that do not bear a T-cell receptor, but can recognize and destroy certain tumour cells and virally infected cells in a manner that is independent of the major histocompatibility complex.

Perforin

A protein that can be released by immune cells and can form pores in the plasma membrane. These pores enable the entrance of serine proteases, such as granzyme B, which initiate caspase cleavage and activation.

Pulsatile and cyclic circumferential stretch

Vascular cells are normally exposed to oscillatory distending and shearing forces owing to the pulsatility of circulating blood. This pulsatility might have an important role in regulating vessel tone and remodelling. Both shear and large-amplitude cyclic stretch have been shown to individually stimulate nitric oxide synthase.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Mehlen, P., Puisieux, A. Metastasis: a question of life or death. Nat Rev Cancer 6, 449–458 (2006). https://doi.org/10.1038/nrc1886

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1886

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