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:

Reprogramming metastatic tumour cells with embryonic microenvironments

Key Points

  • Aggressive tumour cells, such as melanoma, share many characteristics with embryonic progenitors, which contribute to the conundrum of tumour cell plasticity. The challenge is to better understand the aetiology of the plastic, multipotent phenotype and to develop strategies that might include their differentiation and subsequent targeting.

  • A complex and still enigmatic relationship exists between stem cells and their microenvironment that has a crucial role in the determination of cell fate. Current studies identifying the molecular pathways that regulate stem cell plasticity are also examining the epigenetic role of the microenvironment.

  • The microenvironment of human embryonic stem cells can epigenetically reprogramme multipotent metastatic melanoma cells to assume a melanocyte-like phenotype. In addition, the 'reverted' melanoma cells show significantly reduced invasive and tumorigenic ability.

  • The embryonic neural crest microenvironment of the chick provides an attractive model system to explore melanoma tumour cell reprogramming. Human metastatic melanoma cells transplanted into the chick embryonic microenvironment did not form tumours, and a subset of these tumour cells were reprogrammed to a neural crest cell-like phenotype. The melanoma cells also followed neural crest migratory pathways and populated host peripheral structures in a programmed manner.

  • Recent findings using the embryonic zebrafish have illuminated a convergence in the molecular messengers that metastatic tumour and normal stem cells implement during their respective bi-directional communication with the microenvironment, leading to the identification of Nodal.

  • The inhibition of Nodal signalling reduces melanoma cell invasiveness, colony formation and tumorigenicity. Nodal inhibition also promotes the reversion of melanoma cells towards a melanocytic phenotype concomitant with loss of the plastic phenotype.

  • Nodal may represent a new diagnostic marker for disease progression and a novel target for the treatment of aggressive cancers. Additional strategic targets contributing to the Nodal signalling pathway, including SMAD2 and SMAD3, cripto and the activin-like-kinase (ALK) receptor complex, are worth further consideration for inhibiting the plastic tumour cell phenotype.

  • The discovery of key signalling pathways that underlie the commonality of plasticity of embryonic stem cells and multipotent tumour cells will probably result in new therapeutic strategies to suppress the metastatic phenotype.

Abstract

Aggressive tumour cells share many characteristics with embryonic progenitors, contributing to the conundrum of tumour cell plasticity. Recent studies using embryonic models of human stem cells, the zebrafish and the chick have shown the reversion of the metastatic phenotype of aggressive melanoma cells, and revealed the convergence of embryonic and tumorigenic signalling pathways, which may help to identify new targets for therapeutic intervention. This Review will summarize the embryonic models used to reverse the metastatic melanoma phenotype, and highlight the prominent signalling pathways that have emerged as noteworthy targets for future consideration

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: Human embryonic stem cell microenvironment model.
Figure 2: Transplantation of metastatic melanoma cells into the zebrafish embryo.
Figure 3: Embryonic chick model to investigate whether metastatic melanoma cells can be reverted back to their cellular derivatives by the neural crest microenvironment.
Figure 4: The Nodal signalling pathway.

Similar content being viewed by others

References

  1. Topczewska, J. M. et al. Embryonic and tumorigenic pathways converge via Nodal signalling: role in melanoma aggressiveness. Nature Med. 12, 925–932 (2006). This is the first paper to report the discovery of Nodal expression in melanoma and demonstrate its association with tumour cell plasticity and progression.

    CAS  PubMed  Google Scholar 

  2. Sell, S. Stem cell origin of cancer and differentiation therapy. Crit. Rev. Oncol. Hematol. 51, 1–28 (2004).

    PubMed  Google Scholar 

  3. Clarke, M. F. et al. Cancer stem cells – perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res. 66, 9339–9344 (2006).

    CAS  PubMed  Google Scholar 

  4. Li, L. & Neaves, W. B. Normal stem cells and cancer stem cells: The niche matters. Cancer Res. 66, 4553–4557 (2006).

    CAS  PubMed  Google Scholar 

  5. Tan, B. T., Park, C. Y., Ailles, L. E. & Weissman, I. L. The cancer stem cell hypothesis: a work in progress. Lab. Investig. 86, 1203–1207 (2006).

    CAS  PubMed  Google Scholar 

  6. Monk, M. & Holding, C. Human embryonic genes re-expressed in cancer cells. Oncogene 20, 8085–8089 (2001).

    CAS  PubMed  Google Scholar 

  7. Al-Hajj, M., Wicha, M. S., Benito-Hernandez, A., Morrison, S. J. & Clarke, M. F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl Acad. Sci. USA 100, 3983–3988 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Fang, D. et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 65, 9328–9337 (2005).

    CAS  PubMed  Google Scholar 

  9. Houghton, J. et al. Gastric cancer originating from bone marrow-derived cells. Science 306, 1568–1571 (2004).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  11. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001). This paper provides a comprehensive review of stem cells, both normal and cancer.

    CAS  PubMed  Google Scholar 

  12. Singh, S. K. et al. Identification of human brain tumour initiating cells. Nature 432, 396–401 (2004).

    CAS  PubMed  Google Scholar 

  13. Grichnik, J. M. et al. Melanoma, a tumour based on a mutant stem cell? J. Investig. Dermatol. 126, 142–153 (2006).

    CAS  PubMed  Google Scholar 

  14. Virchow, R. L. K. in Cellular Pathology (ed. Hirschwald, A.) (Berlin, 1858).

    Google Scholar 

  15. Garraway, L. A. & Sellers, W. R. Lineage dependency and lineage-survival oncogenes in human cancer. Nature Rev. Cancer 6, 593–602 (2006).

    CAS  Google Scholar 

  16. Mintz, B. & Illmensee, K. Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc. Natl Acad. Sci. USA. 72, 3585–3589 (1975). This seminal study was one of the first to illuminate the ability of embryonic microenvironments to reprogramme tumour cells. Specifically, the embryonic blastocyst microenvironment of the mouse was shown to suppress the tumourigenic phenotype of teratocarcinoma cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Pierce, G. B., Pantazis, C. G., Caldwell, J. E. & Wells, R. S. Specificity of the control of tumour formation by the blastocyst. Cancer Res. 42, 1082–1087 (1982).

    CAS  PubMed  Google Scholar 

  18. Gerschenson, M., Graves, K., Carson, S. D., Wells, R. S. & Pierce, G. B. Regulation of melanoma by the embryonic skin. Proc. Natl Acad. Sci. USA 83, 7307–7310 (1986).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Podesta, A. H., Mullins, J., Pierce, G. B. & Wells, R. S. The neurula stage mouse embryo in control of neuroblastoma. Proc. Natl Acad. Sci. USA 81, 7608–7611 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Dolberg, D. S. & Bissell, M. J. Inability of Rous sarcoma virus to cause sarcomas in the avian embryo. Nature 309, 552–556 (1984).

    CAS  PubMed  Google Scholar 

  21. Bittner, M. et al. Molecular classifcaiton of cutaneous malignant melanoma by gene expression profiling. Nature 406, 536–540 (2000). This was the first report to provide a molecular signature of cutaneous melanoma demonstrating multiple subclasses of the disease.

    CAS  PubMed  Google Scholar 

  22. Carr, K. M., Bittner, M. & Trent, J. M. Gene-expression profiling in human cutaneous melanoma. Oncogene 22, 3076–3080 (2002).

    Google Scholar 

  23. Hoek, K. et al. Expression profiling reveals novel pathways in the transformation of melanocytes to melanoma. Cancer Res. 64, 570–582 (2004).

    Google Scholar 

  24. Gao, C.-F. et al. Proliferation and invasion: plasticity in tumour cells. Proc. Natl Acad. Sci. USA 102, 10528–10533 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Luo, J. et al. Human prostatic cancer and benign prostatic hyperplasia: Molecular dissection by gene expression profiling. Cancer Res. 61, 4683–4688 (2001).

    CAS  PubMed  Google Scholar 

  26. Neve, R. M. et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10, 515–527 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chin, K. et al. Genomic and transcriptional aberrations linked to breast cancer pathologies. Cancer Cell 10, 529–541 (2006).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Lamb, J. et al. The connectivity map: Using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1936 (2006).

    CAS  PubMed  Google Scholar 

  30. Lotem, J. & Sachs, L. Epigenetics and the plasticity of differentiation in normal and cancer stem cells. Oncogene 25, 7663–7672 (2006).

    CAS  PubMed  Google Scholar 

  31. Ivanova, N. B. et al. A stem cell molecular signature. Science 298, 601–604 (2002). This paper used global gene analyses to illuminate the molecular signature of stem cells.

    CAS  PubMed  Google Scholar 

  32. Boyer, L. A. et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122, 947–956 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Maniotis, A. J. et al. Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am. J. Pathol. 155, 739–752 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Maniotis, A. J. et al. Control of melanoma morphogenesis, endothelial survival, and perfusion by extracellular matrix. Lab. Invest. 82, 1031–1034 (2002).

    PubMed  Google Scholar 

  35. Ruf, W. et al. Differential role of tissue factor pathway inhibitors 1 and 2 in melanoma vasculogenic mimicry. Cancer Res. 63, 5381–5389 (2003).

    CAS  PubMed  Google Scholar 

  36. Dome, B., Hendrix, M. J. C., Pahu, S., Tovari, J. & Timar, J. Alternative vascularization mechanisms in cancer: pathology and therapeutic implications. Am. J Pathol. 170, 1–15 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hendrix, M. J. C. et al. Transendothelial function of human metastatic melanoma cells: role of the microenvironment in cell-fate determination. Cancer Res. 62, 665–668 (2002).

    CAS  PubMed  Google Scholar 

  38. Sun, B., Zhang, S., Zhao, X., Zhang, W. & Hao, X. Vasculogenic mimicry is associated with poor survival in patients with mesothelial sarcomas and alveolar rhabdomyosarcomas. Int. J. Oncol. 25, 1609–1614 (2004).

    PubMed  Google Scholar 

  39. Sun, B. et al. Vasculogenic mimicry is associated with high tumour grade, invasion and metastasis, and short survival in patients with hepatocellular carcinoma. Oncol. Rep. 16, 693–698 (2006).

    CAS  PubMed  Google Scholar 

  40. Basu, G. D. et al. A novel role for cyclooxygenase-2 in regulating vascular channel formation by human breast cancer cells. Breast Cancer Res. 8, R69 (2006).

    PubMed  PubMed Central  Google Scholar 

  41. Chung, L. W. et al. Stromal-epithelial interaction in prostate cancer progression. Clin. Genitourin Cancer 5, 162–170 (2006).

    PubMed  Google Scholar 

  42. van der Schaft, D. W. et al. Tumour cell plasticity in Ewing sarcoma, and alternative circulatory system by hypoxia. Cancer Res. 65, 11520–11528 (2005).

    CAS  PubMed  Google Scholar 

  43. Hendrix, M. J. C, Seftor, E. A., Hess, A. R. & Seftor, R. E. B. Vasculogenic mimicry and tumour-cell plasticity: lessons from melanoma. Nature Rev. Cancer 3, 411–421 (2003). This review highlights the molecular underpinnings of melanoma tumour cell plasticity, including vasculogenic mimicry.

    CAS  Google Scholar 

  44. Du, J. et al. MELANA/MART1 and SILV/PMEL17/GP100 are transcriptionally regulated by MITF in melanocytes and melanoma. Am. J. Pathol. 163, 333–343 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Hendrix, M. J. C., Seftor, E. A., Hess, A. R. & Seftor, R. E. B. in From melanocytes to malignant melnoma. (eds Hearing V. J., Leong, S. P. L.) 533–550 (Humana Press, Totowa, 2005).

    Google Scholar 

  46. Takeuchi, H., Kuo, C., Morton, D. L., Wang, H. J. & Hoon, D. S. Expression of differentiation melanoma-associated antigen genes is associated with favorable disease outcome in advanced-stage melanomas. Cancer Res. 63, 441–448 (2003).

    CAS  PubMed  Google Scholar 

  47. Postovit, L. M., Seftor, E. A., Seftor, R. E. B. & Hendrix, M. J. C. A 3-D model to study the epigenetic effects induced by the microenvironment of human embryonic stem cells. Stem Cells 24, 501–505 (2006). This paper describes a new model designed to examine the effects of the microenvironment on cell behaviour. Using this model, the microenvironment of hESCs was shown to reprogramme aggressive melanoma cells towards a less aggressive melanocytic-like phenotype.

    CAS  PubMed  Google Scholar 

  48. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    CAS  PubMed  Google Scholar 

  49. Schatten, G. et al. Culture of human embryonic stem cells. Nature Methods 2, 455–463 (2005).

    CAS  PubMed  Google Scholar 

  50. Levenberg, S. et al. Differentiation of human embryonic stem cells on three-dimensional polymer scaffolds. Proc. Natl Acad. Sci. USA 100, 12741–12746 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Ezashi, T., Das, P. & Roberts, R. M. Low O2 tensions and the prevention of differentiation of hES cells. Proc. Natl Acad. Sci. USA 102, 4783–4788 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Ramalho-Santos, M., et al. 'Stemness': transcriptional profiling of embryonic and adult stem cells. Science 298, 597–600 (2002).

    CAS  PubMed  Google Scholar 

  53. Weissman, I. L. Stem cells: units of development, units of regeneration, and units in evolution. Cell 100, 157–168 (2000).

    CAS  PubMed  Google Scholar 

  54. Silva, G. A. et al. Selective differentiation of neural progenitor cells by high-epitope density nanofibers. Science 303, 1352–1355 (2004).

    CAS  PubMed  Google Scholar 

  55. Xu, R. H. et al. Basic FGF and suppression of BMP signalling sustain undifferentiated proliferation of human ES cells. Nature Methods 2, 185–190 (2005).

    CAS  PubMed  Google Scholar 

  56. Postovit, L.-M. et al. The convergence of embryonic and tumorigenic signalling pathways contribute to tumour cell plasticity. Proceedings ASCB 43, B692 (2006).

    Google Scholar 

  57. Hendrix, M. J., Seftor, E. A., Hess, A. R. & Seftor, R. E. Molecular plasticity of human melanoma cells. Oncogene 19, 3070–3075 (2003).

    Google Scholar 

  58. Balint, K. et al. Activation of Notch1 signalling is required for beta-catenin-mediated human primary melanoma progression. J. Clin. Invest. 115, 3166–3176 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Weeraratna, A. T. et al. Wnt5a signalling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell 1, 279–288 (2002).

    CAS  PubMed  Google Scholar 

  60. De Robertis, E. M., Larrain, J., Oelgeschlager, M. & Wessely, O. The establishment of Spemann's organizer and patterning of the vertebrate embryo. Nature Rev. Genet. 1, 171–181 (2000).

    CAS  PubMed  Google Scholar 

  61. Schier, A. F. Nodal signalling in vertebrate development. Annu. Rev. Cell Dev. Biol. 19, 589–621 (2003). This paper provides an in-depth overview of the Nodal signalling pathway, as well as its function as an embryonic morphogen.

    CAS  PubMed  Google Scholar 

  62. Toyama, R., O'Connell, M. L., Wright, C. V., Kuehn, M. R. & Dawid, I. B. Nodal induces ectopic goosecoid and lim1 expression and axis duplication in zebrafish. Development 121, 383–391 (1995).

    CAS  PubMed  Google Scholar 

  63. Thisse, B., Wright, C. V. E. & Thisse, C. Activin- and Nodal-related factors control antero–posterior patterning of the zebrafish embryo. Nature 403, 425–428 (2000).

    CAS  PubMed  Google Scholar 

  64. Cucina, A. et al. Zebrafish embryo proteins induce apoptosis in human colon cancer cells (Caco2). Apoptosis 11, 1617–1628 (2006).

    CAS  PubMed  Google Scholar 

  65. Lee, L. M., Seftor, E. A., Bonde, G., Cornell, R. A. & Hendrix, M. J. The fate of human malignant melanoma cells transplanted into zebrafish embryos: assessment of migration and cell division in the absence of tumour formation. Dev. Dyn. 233, 1560–1570 (2005).

    CAS  PubMed  Google Scholar 

  66. Haldi, M., Ton, C., Seng, W. L. & McGrath, P. Human melanoma cells transplanted into zebrafish proliferate, migrate, produce melanin, form masses and stimulate angiogenesis in zebrafish. Angiogenesis 9, 139–151 (2006).

    PubMed  Google Scholar 

  67. LeDouarin, N. & Kalcheim, C. The Neural Crest. Cambridge Univ. Press, Cambridge, UK (1999).

    Google Scholar 

  68. Trainor, P. & Krumlauf, R. Patterning the cranial neural crest: hindbrain segmentation and Hox gene plasticity. Nature Rev. Neuro. 1, 116–124 (2000).

    CAS  Google Scholar 

  69. Kontges, G. & Lumsden, A. Rhombencephalic neural crest segmentation is preserved throughout craniofacial ontogeny. Development 122, 3229–3242 (1996).

    CAS  PubMed  Google Scholar 

  70. Dupin, E. & LeDouarin, N. Development of melanocyte precursors from the vertebrate neural crest. Oncogene 22, 3016–3023 (2003)

    CAS  PubMed  Google Scholar 

  71. Harris, M. & Erickson, C. Lineage specification in neural crest cell pathfinding. Dev. Dyn. 236, 1–19 (2007).

    CAS  PubMed  Google Scholar 

  72. Noden, D. The role of the neural crest in patterning of avian cranial skeletal, connective, and muscle tissues. Dev. Biol. 96, 144–165 (1983).

    CAS  PubMed  Google Scholar 

  73. Lumsden, A., Sprawson, N. & Graham, A. Segmental origin and migration of neural crest cells in the hindbrain region of the chick embryo. Development 113, 1281–1291 (1991).

    CAS  PubMed  Google Scholar 

  74. Trainor, P. & Krumlauf, R. Plasticity in mouse neural crest cells reveals a new patterning role for cranial mesoderm. Nature Cell Biol. 2, 96–102 (2000).

    CAS  PubMed  Google Scholar 

  75. Graham, A., Heyman, I. & Lumsden, A. Even-numbered rhombomeres control the apoptotic elimination of neural crest cells from odd-numbered rhombomeres in the chick hindbrain. Development 119, 233–245 (1993).

    CAS  PubMed  Google Scholar 

  76. Grapin-Botton, A. et al. Plasticity of transposed rhombomeres: Hox gene induction is correlated with phenotypic modifications. Development 121, 2707–2721 (1995).

    CAS  PubMed  Google Scholar 

  77. Schilling, T., Prince, V. & Ingham, P. Plasticity in zebrafish Hox expression in the hindbrain and cranial neural crest. Dev. Biol. 231, 201–216 (2001).

    CAS  PubMed  Google Scholar 

  78. Kulesa, P. & Fraser, S. Neural crest cell dynamics revealed by time-lapse video microscopy of whole chick explant cultures. Dev. Biol. 204, 327–344 (1998).

    CAS  PubMed  Google Scholar 

  79. Schilling, T. & Kimmel, C. Segment and cell type lineage restrictions during pharyngeal arch development in the zebrafish embryo. Development 120, 483–494 (1994).

    CAS  PubMed  Google Scholar 

  80. Smith, A. et al. The EphA4 and EphB1 receptor tyrosine kinases and ephrin-B2 ligand regulate targeted migration of branchial neural crest cells. Curr. Biol. 7, 561–570 (1997).

    CAS  PubMed  Google Scholar 

  81. Farlie, P. et al. A paraxial exclusion zone creates patterned cranial neural crest cell outgrowth adjacent to rhombomeres 3 and 5. Dev. Biol. 213, 70–84 (1999).

    CAS  PubMed  Google Scholar 

  82. Kulesa, P. et al. Reprogramming metastatic melanoma cells to assume a neural crest cell-like phenotype in an embryonic microenvironment. Proc. Natl Acad. Sci. USA 103, 3752–3757 (2006). This paper illustrates the ability of the chick embryonic neural crest to reprogramme metastatic melanoma cells towards neural-crest associated phenotypes.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Real, C. et al. Clonally cultured differentiated pigment cells can dedifferentiate and generate multipotent progenitors with self-renewal potential. Dev. Biol. 300, 656–669 (2006).

    CAS  PubMed  Google Scholar 

  84. Erickson, C., Tosney, K. & Weston, J. Analysis of migratory behavior of neural crest and fibroblastic cells in embryonic tissues. Dev. Biol. 77, 142–156 (1980).

    CAS  PubMed  Google Scholar 

  85. Oppitz, M. et al. Non-malignant migration of B16 mouse melanoma cells in the neural crest and invasive growth in the eye cup of the chick embryo. Melanoma Res. 1, 17–30 (2007).

    Google Scholar 

  86. Teddy, J. & Kulesa, P. In vivo evidence for short- and long-range cell communication in cranial neural crest cells. Development 131, 6141–6151 (2004).

    CAS  PubMed  Google Scholar 

  87. Young, H. et al. Dynamics of neural crest-derived cell migration in the embryonic mouse gut. Dev. Biol. 270, 455–473 (2004).

    CAS  PubMed  Google Scholar 

  88. Drukenbrod, N. & Epstein, M. Behavior of enteric neural crest-derived cells varies with respect to the migratory wavefront. Dev. Dyn. 236, 84–92 (2007).

    Google Scholar 

  89. Chen, C. & Shen, M. M. Two modes by which Lefty proteins inhibit Nodal signalling. Curr. Biol. 14, 618–624.

  90. Postovit, L. M. et al. The commonality of plasticity underlying multipotent tumour cells and embryonic stem cells. J. Cell Biochem. 19 December 2006 [Epub ahead of print].

  91. The Cancer Genome Anatomy Project. SAGE Anatomic Viewer. CGAP [online], (2007).

  92. Mesnard, D., Guzman-Ayala, M. & Constam, D. B. Nodal specifies embryonic visceral endoderm and sustains pluripotent cells in the epiblast before overt axial patterning. Development 133, 2497–2505 (2006).

    CAS  PubMed  Google Scholar 

  93. Jager, D., Jager, E. & Knuth, A. Immune response to tumour antigens: implications for antigen specific immunotherapy of cancer. J. Clin. Pathol. 54, 669–673 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Gibbs, W. W. Nanobodies. Sci. Am. 293, 78–83 (2005).

    PubMed  Google Scholar 

  95. Strizzi, L., Bianco, C., Normanno, N. & Salomon, D. Cripto-1: A multifunciontal modulator during embryogenesis and angiogenesis. Oncogene 24, 5731–5741 (2005).

    CAS  PubMed  Google Scholar 

  96. Minchiotti, G. Nodal-dependant Cripto signalling in ES cells: from stem cells to tumour biology. Oncogene 24, 5668–5675 (2005).

    CAS  PubMed  Google Scholar 

  97. Bianco, C. et al. Identification of cripto-1 as a novel serologic marker for breast and colon cancer. Clin. Cancer Res. 12, 5158–5164 (2006).

    CAS  PubMed  Google Scholar 

  98. Strizzi, L. et al. Epithelial mesenchymal transition is a characteristic of hyperplasias and tumors in mammary gland from MMTV-Cripto-1 transgenic mice. J. Cell Physiol. 201, 266–276 (2004).

    CAS  PubMed  Google Scholar 

  99. Normanno, N. et al. Cripto-1 overexpression leads to enhanced invasiveness and resistance to anoikis in human MCF-7 breast cancer cells. J. Cell Physiol. 198, 31–39 (2004).

    CAS  PubMed  Google Scholar 

  100. Brandt, R. et al. Identification and biological characterization of an epidermal growth factor-related protein: cripto-1. J. Biol. Chem. 269, 17320–17328 (1994).

    CAS  PubMed  Google Scholar 

  101. Bianco, C. et al. A nodal- and ALK4-independent signalling pathway activated by Cripto-1 through glypican-1 and c-Src. Cancer Res. 63, 1192–1197 (2003).

    CAS  PubMed  Google Scholar 

  102. Adkins, H. B. et al. Antibody blockade of the Cripto CFC domain suppresses tumour cell growth in vivo. J. Clin. Invest. 112, 575–587 (2003). This paper demonstrates the importance of targeting Cripto on cancer cells as a rational therapeutic strategy.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Xing, P. X., Hu, X. F., Pietersz, G. A., Hosick, H. L. & McKenzie, I. F. Cripto: a novel target for antibody-based cancer immunotherapy. Cancer Res. 64, 4018–4023 (2004).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to gratefully acknowledge the help of J. Topczewska, J. Topczewski, N. Margaryan, A. Hess and B. Nickoloff. This research was supported by a grant from the National Cancer Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mary J. C. Hendrix.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

National Cancer Institute

FURTHER INFORMATION

Mary Hendrix's homepage

Glossary

Tumour cell plasticity

The ability of aggressive tumour cells to express multiple molecular phenotypes similar to pluripotent, embryonic-like stem cells.

Neovascularization

A formation of functional microvascular networks with red blood cell perfusion that differs from angiogenesis, which is characterized by the protrusion and outgrowth of capillary buds and sprouts from pre-existing blood vessels.

Synoviosarcoma

A malignant neoplasm arising in the synovial membrane of the joints and in the synovial cells of tendons and bursae; also called malignant synovioma and synovial sarcoma.

Phaeochromocytoma

A tumour that forms in the centre of the adrenal gland that causes it to make too much adrenaline.

Ewings sarcoma

A highly malignant, metastatic, small round-cell tumour of the bone that usually occurs in the diaphyses (shafts) of long bones, ribs and flat bones of children or adolescents.

Spheroid

A spherical aggregation of tumour cells, grown in tissue culture, that reflects many of the properties of solid tumours. Spheroids have been used for studying the penetration of anticancer drugs into tumour tissue.

Amelanotic

A complete lack of the pigment melanin in pigment-derived cells and tissues.

Feeder-free matrices

The growth of embryonic stem cells in vitro in the absence of an underlying layer of mouse-derived fibroblasts.

Animal pole

The point in the blastocyst that is farthest away from the yoke margin. In the zebrafish, mesoendoderm is not usually found here.

Dorsal organizer

A group of cells on the dorsal lip of the blastopore that induces the differentiation of cells in the embryo, controlling the growth and development of adjacent parts that eventually form the body axis.

Neural tube

In the developing vertebrate nervous system, the neural tube is the precursor of the central nervous system, which comprises the brain and spinal cord.

Embryonic axis

An imaginary line from the head end to the tail end of an embryo or, before that, the line of elongation of the primitive streak and groove.

Filopodial extensions

Filopodia are slender cytoplasmic projections that extend from the leading edge of migrating cells and contain actin filaments crosslinked into bundles by actin-binding proteins. They form focal adhesions that function to link the cell surface to the substratum and facilitate cell motility.

Morpholinos

Morpholino oligos are short chains of about 25 Morpholino subunits comprised of a nucleic acid base, a morpholine ring and a non–ionic phosphorodiamidate intersubunit linkage. Their high mRNA-binding affinity and specificity permits them to sterically block translation initiation in the cytosol, modify pre-mRNA splicing in the nucleus or directly block miRNA activity to effectively knock down the expression of targeted genes.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hendrix, M., Seftor, E., Seftor, R. et al. Reprogramming metastatic tumour cells with embryonic microenvironments. Nat Rev Cancer 7, 246–255 (2007). https://doi.org/10.1038/nrc2108

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc2108

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