Skip to main content

Advertisement

Log in

Tumour vascularization: sprouting angiogenesis and beyond

  • Published:
Cancer and Metastasis Reviews Aims and scope Submit manuscript

Abstract

Tumour angiogenesis is a fast growing domain in tumour biology. Many growth factors and mechanisms have been unravelled. For almost 30 years, the sprouting of new vessels out of existing ones was considered as an exclusive way of tumour vascularisation. However, over the last years several additional mechanisms have been identified. With the discovery of the contribution of intussusceptive angiogenesis, recruitment of endothelial progenitor cells, vessel co-option, vasculogenic mimicry and lymphangiogenesis to tumour growth, anti-tumour targeting strategies will be more complex than initially thought. This review highlights these processes and intervention as a potential application in cancer therapy. It is concluded that future anti-vascular therapies might be most beneficial when based on multimodal anti-angiogenic, anti-vasculogenic mimicry and anti-lymphangiogenic strategies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Carmeliet, P., & Jain, R. K. (2000). Angiogenesis in cancer and other diseases. Nature, 407, 249–57.

    PubMed  CAS  Google Scholar 

  2. Martin, A., Komada, M. R., & Sane, D. C. (2003). Abnormal angiogenesis in diabetes mellitus. Medicinal Research Reviews, 23, 117–45.

    PubMed  CAS  Google Scholar 

  3. Koch, A. E. (2003). Angiogenesis as a target in rheumatoid arthritis. Annals of the Rheumatic Diseases, 62 Suppl 2, ii60–7.

    PubMed  CAS  Google Scholar 

  4. Cao, Y., Hong, A., Schulten, H., & Post, M. J. (2005). Update on therapeutic neovascularization. Cardiovascular Research, 65, 639–48.

    PubMed  CAS  Google Scholar 

  5. Carmeliet, P. (2005). Angiogenesis in life, disease and medicine. Nature, 438, 932–36.

    PubMed  CAS  Google Scholar 

  6. Hanahan, D., & Weinberg, R. A. (2000). The hallmarks of cancer. Cell, 100, 57–0.

    PubMed  CAS  Google Scholar 

  7. Ferrara, N., Gerber, H. P., & LeCouter, J. (2003). The biology of VEGF and its receptors. Nature Medicine, 9, 669–76

    PubMed  CAS  Google Scholar 

  8. Jain, R. K. (2003). Molecular regulation of vessel maturation. Nature Medicine, 9, 685–93.

    PubMed  CAS  Google Scholar 

  9. Kerbel, R. S. (2000). Tumor angiogenesis: Past, present and the near future. Carcinogenesis, 21, 505–15.

    PubMed  CAS  Google Scholar 

  10. Itoh, N., & Ornitz, D. M. (2004). Evolution of the Fgf and Fgfr gene families. Trends in Genetics, 20, 563–69.

    PubMed  CAS  Google Scholar 

  11. Presta, M., Dell’Era, P., Mitola, S., Moroni, E., Ronca, R., & Rusnati, M. (2005). Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Reviews, 16, 159–78.

    PubMed  CAS  Google Scholar 

  12. Gross, J. L., Herblin, W. F., Dusak, B. A., Czerniak, P., Diamond, M. D., Sun, T., et al. (1993). Effects of modulation of basic fibroblast growth factor on tumor growth in vivo. Journal of the National Cancer Institute, 85, 121–31.

    PubMed  Google Scholar 

  13. Wang, Y. & Becker, D. (1997). Antisense targeting of basic fibroblast growth factor and fibroblast growth factor receptor-1 in human melanomas blocks intratumoral angiogenesis and tumor growth. Nature Medicine, 3, 887–93.

    PubMed  CAS  Google Scholar 

  14. Ferrara, N., & Davis-Smyth, T. (1997). The biology of vascular endothelial growth factor. Endocrine Reviews, 18, 4–5.

    PubMed  CAS  Google Scholar 

  15. Bergers, G., Brekken, R., McMahon, G., Vu, T. H., Itoh, T., Tamaki, K., et al. (2000). Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nature Cell Biology, 2, 737–44.

    PubMed  CAS  Google Scholar 

  16. Iyer, S., & Acharya, K. R. (2002). Role of placenta growth factor in cardiovascular health. Trends in Cardiovascular Medicine, 12, 128–34.

    PubMed  CAS  Google Scholar 

  17. Carmeliet, P., Moons, L., Luttun, A., Vincenti, V., Compernolle, V., De Mol, M., et al. (2001). G. Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nature Medicine, 7, 575–83.

    PubMed  CAS  Google Scholar 

  18. Davis, S., Aldrich, T. H., Jones, P. F., Acheson, A., Compton, D. L., Jain, V., et al. (1996). Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning. Cell, 87, 1161–169.

    PubMed  CAS  Google Scholar 

  19. Maisonpierre, P. C., Suri, C., Jones, P. F., Bartunkova, S., Wiegand, S. J., Radziejewski, C., et al. (1997). Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science, 277, 55–0.

    PubMed  CAS  Google Scholar 

  20. Elliott, R. L., & Blobe, G. C. (2005). Role of transforming growth factor Beta in human cancer. Journal of Clinical Oncology, 23, 2078–093.

    PubMed  CAS  Google Scholar 

  21. Armulik, A., Abramsson, A., & Betsholtz, C. (2005). Endothelial/pericyte interactions. Circulation Research, 97, 512–23.

    PubMed  CAS  Google Scholar 

  22. Petit, A. M., Rak, J., Hung, M. C., Rockwell, P., Goldstein, N., Fendly, B., et al. (1997). Neutralizing antibodies against epidermal growth factor and ErbB-2/neu receptor tyrosine kinases down-regulate vascular endothelial growth factor production by tumor cells in vitro and in vivo: angiogenic implications for signal transduction therapy of solid tumors. American Journal of Pathology, 151, 1523–530.

    PubMed  CAS  Google Scholar 

  23. Li, A., Dubey, S., Varney, M. L., Dave, B. J., & Singh, R. K. (2003). IL-8 directly enhanced endothelial cell survival, proliferation, and matrix metalloproteinases production and regulated angiogenesis. Journal of Immunology, 170, 3369–376.

    CAS  Google Scholar 

  24. Gerhardt, H., Golding, M., Fruttiger, M., Ruhrberg, C., Lundkvist, A., Abramsson, A., et al. (2003). VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. Journal of Cell Biology, 161, 1163–177.

    PubMed  CAS  Google Scholar 

  25. Brantley, D. M., Cheng, N., Thompson, E. J., Lin, Q., Brekken, R. A., Thorpe, P. E., et al. (2002). Soluble Eph A receptors inhibit tumor angiogenesis and progression in vivo. Oncogene, 21, 7011–026.

    PubMed  CAS  Google Scholar 

  26. Bielenberg, D. R., Hida, Y., Shimizu, A., Kaipainen, A., Kreuter, M., Kim, C. C., et al. (2004). Semaphorin 3F, a chemorepulsant for endothelial cells, induces a poorly vascularized, encapsulated, nonmetastatic tumor phenotype. Journal of Clinical Investigation, 114, 1260–271.

    PubMed  CAS  Google Scholar 

  27. Conrotto, P., Valdembri, D., Corso, S., Serini, G., Tamagnone, L., Comoglio, P. M., et al. (2005). Sema4D induces angiogenesis through Met recruitment by Plexin B1. Blood, 105, 4321–329.

    PubMed  CAS  Google Scholar 

  28. Wang, B., Xiao, Y., Ding, B. B., Zhang, N., Yuan, X., Gui, L., et al. (2003). Induction of tumor angiogenesis by Slit-Robo signaling and inhibition of cancer growth by blocking Robo activity. Cancer Cell, 4, 19–9.

    PubMed  Google Scholar 

  29. Arakawa, H. (2004). Netrin-1 and its receptors in tumorigenesis. Natural Reviews Cancer, 4, 978–87.

    CAS  Google Scholar 

  30. Rehman, A. O., & Wang, C. Y. (2006). Notch signaling in the regulation of tumor angiogenesis. Trends in Cell Biology, 16, 293–00.

    PubMed  CAS  Google Scholar 

  31. Rastinejad, F., Polverini, P. J., & Bouck, N. P. (1989). Regulation of the activity of a new inhibitor of angiogenesis by a cancer suppressor gene. Cell, 56, 345–55.

    PubMed  CAS  Google Scholar 

  32. Ferrara, N., Clapp, C., & Weiner, R. (1991). The 16K fragment of prolactin specifically inhibits basal or fibroblast growth factor stimulated growth of capillary endothelial cells. Endocrinology, 129, 896–00.

    PubMed  CAS  Google Scholar 

  33. Kolber, D. L., Knisely, T. L., and Maione, T. E. (1995). Inhibition of development of murine melanoma lung metastases by systemic administration of recombinant platelet factor 4. Journal of National Cancer Institute, 87, 304–09.

    PubMed  CAS  Google Scholar 

  34. Luster, A. D., Greenberg, S. M., & Leder, P. (1995). The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial cell proliferation. Journal of Experimental Medicine, 182, 219–31.

    PubMed  CAS  Google Scholar 

  35. O’Reilly, M. S., Holmgren, L., Shing, Y., Chen, C., Rosenthal, R. A., Moses, M., et al. (1994). Angiostatin: A novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell, 79, 315–28.

    PubMed  Google Scholar 

  36. O’Reilly, M. S., Boehm, T., Shing, Y., Fukai, N., Vasios, G., Lane, W. S., et al. (1997). Endostatin: An endogenous inhibitor of angiogenesis and tumor growth. Cell, 88, 277–85.

    PubMed  Google Scholar 

  37. van der Schaft, D. W., Toebes, E. A., Haseman, J. R., Mayo, K. H., & Griffioen, A. W. (2000). Bactericidal/permeability-increasing protein (BPI) inhibits angiogenesis via induction of apoptosis in vascular endothelial cells. Blood, 96, 176–81.

    PubMed  Google Scholar 

  38. Maeshima, Y., Colorado, P. C., Torre, A., Holthaus, K. A., Grunkemeyer, J. A., Ericksen, M. B., et al. (2000). Distinct antitumor properties of a type IV collagen domain derived from basement membrane. Journal of Biological Chemistry, 275, 21340–1348.

    PubMed  CAS  Google Scholar 

  39. Tabruyn, S. P., & Griffioen, A. W. (2007). Molecular pathways of angiogenesis inhibition. Biochemical and Biophysical Research Communications, 355, 1–.

    PubMed  CAS  Google Scholar 

  40. Marx, J. Angiogenesis. (2003). A boost for tumor starvation. Science, 301, 452–54.

    PubMed  CAS  Google Scholar 

  41. Ferrara, N., & Kerbel, R. S. (2005). Angiogenesis as a therapeutic target. Nature, 438, 967–74.

    PubMed  CAS  Google Scholar 

  42. Hurwitz, H., Fehrenbacher, L., Novotny, W., Cartwright, T., Hainsworth, J., Heim, W., et al. (2004). Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. New England Journal of Medicine, 350, 2335–342.

    PubMed  CAS  Google Scholar 

  43. Glusker, P., Recht, L., and Lane, B. (2006). Reversible posterior leukoencephalopathy syndrome and bevacizumab. New England Journal of Medicine, 354, 980–82; discussion 980–82.

    PubMed  CAS  Google Scholar 

  44. Ozcan, C., Wong, S. J., and Hari, P. (2006). Reversible posterior leukoencephalopathy syndrome and bevacizumab. New England Journal of Medicine, 354, 980–82, discussion 980–82.

    PubMed  Google Scholar 

  45. Alitalo, K., Tammela, T., & Petrova, T. V. (2005). Lymphangiogenesis in development and human disease. Nature, 438, 946–53.

    PubMed  CAS  Google Scholar 

  46. Yu, J. L., Rak, J. W., Coomber, B. L., Hicklin, D. J., & Kerbel, R. S. (2002). Effect of p53 status on tumor response to antiangiogenic therapy. Science, 295, 1526–528.

    PubMed  CAS  Google Scholar 

  47. Glade Bender, J., Cooney, E. M., Kandel, J. J., & Yamashiro, D. J. (2004). Vascular remodeling and clinical resistance to antiangiogenic cancer therapy. Drug Resistance Updates, 7, 289–00.

    PubMed  CAS  Google Scholar 

  48. Hida, K., Hida, Y., Amin, D. N., Flint, A. F., Panigrahy, D., Morton, C. C., et al. (2004). Tumor-associated endothelial cells with cytogenetic abnormalities. Cancer Research, 64, 8249–255.

    PubMed  CAS  Google Scholar 

  49. Streubel, B., Chott, A., Huber, D., Exner, M., Jager, U., Wagner, O., et al. (2004). Lymphoma-specific genetic aberrations in microvascular endothelial cells in B-cell lymphomas. New England Journal of Medicine, 351, 250–59.

    PubMed  CAS  Google Scholar 

  50. Caduff, J. H., Fischer, L. C., & Burri, P. H. (1986). Scanning electron microscope study of the developing microvasculature in the postnatal rat lung. Anatomical Record, 216, 154–64.

    PubMed  CAS  Google Scholar 

  51. Burri, P. H., Hlushchuk, R., & Djonov, V. (2004). Intussusceptive angiogenesis: Its emergence, its characteristics, and its significance. Developmental Dynamics, 231, 474–88.

    PubMed  Google Scholar 

  52. Djonov, V. G., Kurz, H., & Burri, P. H. (2002). Optimality in the developing vascular system: branching remodeling by means of intussusception as an efficient adaptation mechanism. Developmental Dynamics, 224, 391–02.

    PubMed  Google Scholar 

  53. Patan, S., Haenni, B., & Burri, P. H. (1993). Evidence for intussusceptive capillary growth in the chicken chorio-allantoic membrane (CAM). Anatomy and Embryology (Berl), 187, 121–30.

    CAS  Google Scholar 

  54. Patan, S., Munn, L. L., & Jain, R. K. (1996). Intussusceptive microvascular growth in a human colon adenocarcinoma xenograft: A novel mechanism of tumor angiogenesis. Microvascular Research, 51, 260–72.

    PubMed  CAS  Google Scholar 

  55. Djonov, V., Andres, A. C., & Ziemiecki, A. (2001). Vascular remodelling during the normal and malignant life cycle of the mammary gland. Microscopy Research and Technique, 52, 182–89.

    PubMed  CAS  Google Scholar 

  56. Ribatti, D., Nico, B., Floris, C., Mangieri, D., Piras, F., Ennas, M. G., et al. (2005). Microvascular density, vascular endothelial growth factor immunoreactivity in tumor cells, vessel diameter and intussusceptive microvascular growth in primary melanoma. Oncology Reports, 14, 81–4.

    PubMed  Google Scholar 

  57. Oh, S. J., Kurz, H., Christ, B., & Wilting, J. (1998). Platelet-derived growth factor-B induces transformation of fibrocytes into spindle-shaped myofibroblasts in vivo. Histochemistry and Cell Biology, 109, 349–57.

    PubMed  CAS  Google Scholar 

  58. Thurston, G., Suri, C., Smith, K., McClain, J., Sato, T. N., Yancopoulos, G. D., et al. (1999). Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1. Science, 286, 2511–514.

    PubMed  CAS  Google Scholar 

  59. Asahara, T., Murohara, T., Sullivan, A., Silver, M., van der Zee, R., Li, T., et al. (1997). Isolation of putative progenitor endothelial cells for angiogenesis. Science, 275, 964–67.

    PubMed  CAS  Google Scholar 

  60. Kalka, C., Masuda, H., Takahashi, T., Kalka-Moll, W. M., Silver, M., Kearney, M., et al. (2000). Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proceedings of the National Academy of Sciences of the United States of America, 3422–427.

  61. Lin, Y., Weisdorf, D. J., Solovey, A., & Hebbel, R. P. (2000). Origins of circulating endothelial cells and endothelial outgrowth from blood. Journal of Clinical Investigation, 105, 71–7.

    PubMed  CAS  Google Scholar 

  62. Heissig, B., Hattori, K., Dias, S., Friedrich, M., Ferris, B., Hackett, N. R., et al. (2002). Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell, 109, 625–37.

    PubMed  CAS  Google Scholar 

  63. Asahara, T., Takahashi, T., Masuda, H., Kalka, C., Chen, D., Iwaguro, H., et al. (1999). VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO Journal, 18, 3964–972.

    PubMed  CAS  Google Scholar 

  64. Hattori, K., Heissig, B., Wu, Y., Dias, S., Tejada, R., Ferris, B., et al. (2002). Placental growth factor reconstitutes hematopoiesis by recruiting VEGFR1(+) stem cells from bone-marrow microenvironment. Nature Medicine, 8, 841–49.

    PubMed  CAS  Google Scholar 

  65. Moore, M. A., Hattori, K., Heissig, B., Shieh, J. H., Dias, S., Crystal, R. G., and Rafii, S. (2001). Mobilization of endothelial and hematopoietic stem and progenitor cells by adenovector-mediated elevation of serum levels of SDF-1, VEGF, and angiopoietin-1. Annals of the New York Academy of Sciences, 938, 36–5; discussion 45–7.

    Article  PubMed  CAS  Google Scholar 

  66. Aicher, A., Heeschen, C., Mildner-Rihm, C., Urbich, C., Ihling, C., Technau-Ihling, K., et al. (2003). Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nature Medicine, 9, 1370–376.

    PubMed  CAS  Google Scholar 

  67. Takahashi, T., Kalka, C., Masuda, H., Chen, D., Silver, M., Kearney, M., et al. (1999). Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nature Medicine, 5, 434–38.

    PubMed  CAS  Google Scholar 

  68. Vajkoczy, P., Blum, S., Lamparter, M., Mailhammer, R., Erber, R., Engelhardt, B., et al. (2003). Multistep nature of microvascular recruitment of ex vivo-expanded embryonic endothelial progenitor cells during tumor angiogenesis. Journal of Experimental Medicine, 197, 1755–765.

    PubMed  CAS  Google Scholar 

  69. Deb, A., Skelding, K. A., Wang, S., Reeder, M., Simper, D., & Caplice, N. M. (2004). Integrin profile and in vivo homing of human smooth muscle progenitor cells. Circulation, 110, 2673–677.

    PubMed  CAS  Google Scholar 

  70. Chavakis, E., Hain, A., Vinci, M., Carmona, G., Bianchi, M. E., Vajkoczy, P., et al. (2007). High-mobility group box 1 activates integrin-dependent homing of endothelial progenitor cells. Circulation Research, 100, 204–12.

    PubMed  CAS  Google Scholar 

  71. Liao, F., Huynh, H. K., Eiroa, A., Greene, T., Polizzi, E., & Muller, W. A. (1995). Migration of monocytes across endothelium and passage through extracellular matrix involve separate molecular domains of PECAM-1. Journal of Experimental Medicine, 182, 1337–343.

    PubMed  CAS  Google Scholar 

  72. Schenkel, A. R., Mamdouh, Z., Chen, X., Liebman, R. M., & Muller, W. A. (2002). CD99 plays a major role in the migration of monocytes through endothelial junctions. Nature Immunology, 3, 143–50.

    PubMed  CAS  Google Scholar 

  73. Gehling, U. M., Ergun, S., Schumacher, U., Wagener, C., Pantel, K., Otte, M., et al. (2000). In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood, 95, 3106–112.

    PubMed  CAS  Google Scholar 

  74. Asahara, T., Masuda, H., Takahashi, T., Kalka, C., Pastore, C., Silver, M., et al. (1999). Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circulation Research, 85, 221–28.

    PubMed  CAS  Google Scholar 

  75. Lyden, D., Young, A. Z., Zagzag, D., Yan, W., Gerald, W., O’Reilly, R., et al. (1999). Id1 and Id3 are required for neurogenesis, angiogenesis and vascularization of tumour xenografts. Nature, 401, 670–77.

    PubMed  CAS  Google Scholar 

  76. Lyden, D., Hattori, K., Dias, S., Costa, C., Blaikie, P., Butros, L., et al. (2001). Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nature Medicine, 7, 1194–201.

    PubMed  CAS  Google Scholar 

  77. Machein, M. R., Renninger, S., de Lima-Hahn, E., & Plate, K. H. (2003). Minor contribution of bone marrow-derived endothelial progenitors to the vascularization of murine gliomas. Brain Pathology, 13, 582–97.

    Article  PubMed  CAS  Google Scholar 

  78. Ruzinova, M. B., Schoer, R. A., Gerald, W., Egan, J. E., Pandolfi, P. P., Rafii, S., et al. (2003). Effect of angiogenesis inhibition by Id loss and the contribution of bone-marrow-derived endothelial cells in spontaneous murine tumors. Cancer Cell, 4, 277–89.

    PubMed  CAS  Google Scholar 

  79. Gothert, J. R., Gustin, S. E., van Eekelen, J. A., Schmidt, U., Hall, M. A., Jane, S. M., et al. (2004). Genetically tagging endothelial cells in vivo: Bone marrow-derived cells do not contribute to tumor endothelium. Blood, 104, 1769–777.

    PubMed  Google Scholar 

  80. Rajantie, I., Ilmonen, M., Alminaite, A., Ozerdem, U., Alitalo, K., & Salven, P. (2004). Adult bone marrow-derived cells recruited during angiogenesis comprise precursors for periendothelial vascular mural cells. Blood, 104, 2084–086.

    PubMed  CAS  Google Scholar 

  81. Peters, B. A., Diaz, L. A., Polyak, K., Meszler, L., Romans, K., Guinan, E. C., et al. (2005). Contribution of bone marrow-derived endothelial cells to human tumor vasculature. Nature Medicine, 11, 261–62.

    PubMed  CAS  Google Scholar 

  82. Larrivee, B., Niessen, K., Pollet, I., Corbel, S. Y., Long, M., Rossi, F. M., et al. (2005). Minimal contribution of marrow-derived endothelial precursors to tumor vasculature. Journal of Immunology, 175, 2890–899.

    CAS  Google Scholar 

  83. Sussman, L. K., Upalakalin, J. N., Roberts, M. J., Kocher, O., & Benjamin, L. E. (2003). Blood markers for vasculogenesis increase with tumor progression in patients with breast carcinoma. Cancer Biology & Therapy, 2, 255–56.

    CAS  Google Scholar 

  84. Kim, H. K., Song, K. S., Kim, H. O., Chung, J. H., Lee, K. R., Lee, Y. J., et al. (2003). Circulating numbers of endothelial progenitor cells in patients with gastric and breast cancer. Cancer Letter, 198, 83–8.

    CAS  Google Scholar 

  85. Capillo, M., Mancuso, P., Gobbi, A., Monestiroli, S., Pruneri, G., Dell’Agnola, C., et al. (2003). Continuous infusion of endostatin inhibits differentiation, mobilization, and clonogenic potential of endothelial cell progenitors. Clinical Cancer Research, 9, 377–82.

    PubMed  CAS  Google Scholar 

  86. Shaked, Y., Bertolini, F., Man, S., Rogers, M. S., Cervi, D., Foutz, T., et al. (2005). Genetic heterogeneity of the vasculogenic phenotype parallels angiogenesis; Implications for cellular surrogate marker analysis of antiangiogenesis. Cancer Cell, 7, 101–11.

    PubMed  CAS  Google Scholar 

  87. Arafat, W. O., Casado, E., Wang, M., Alvarez, R. D., Siegal, G. P., Glorioso, J. C., et al. (2000). Genetically modified CD34+ cells exert a cytotoxic bystander effect on human endothelial and cancer cells. Clinical Cancer Research, 6, 4442–448.

    PubMed  CAS  Google Scholar 

  88. Ingram, D. A., Caplice, N. M., & Yoder, M. C. (2005). Unresolved questions, changing definitions, and novel paradigms for defining endothelial progenitor cells. Blood, 106, 1525–531.

    PubMed  CAS  Google Scholar 

  89. Wesseling, P., van der Laak, J. A., de Leeuw, H., Ruiter, D. J., & Burger, P. C. (1994). Quantitative immunohistological analysis of the microvasculature in untreated human glioblastoma multiforme. Computer-assisted image analysis of whole-tumor sections. Journal of Neurosurgery, 81, 902–09.

    PubMed  CAS  Google Scholar 

  90. Holmgren, L., O’Reilly, M. S., & Folkman, J. (1995). Dormancy of micrometastases: Balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nature Medicine, 1, 149–53.

    PubMed  CAS  Google Scholar 

  91. Pezzella, F., Pastorino, U., Tagliabue, E., Andreola, S., Sozzi, G., Gasparini, G., et al. (1997). Non-small-cell lung carcinoma tumor growth without morphological evidence of neo-angiogenesis. American Journal of Pathology, 151, 1417–423.

    PubMed  CAS  Google Scholar 

  92. Holash, J., Maisonpierre, P. C., Compton, D., Boland, P., Alexander, C. R., Zagzag, D., et al. (1999). Vessel co-option, regression, and growth in tumors mediated by angiopoietins and VEGF. Science, 284, 1994–998.

    PubMed  CAS  Google Scholar 

  93. Zhang, L., Yang, N., Park, J. W., Katsaros, D., Fracchioli, S., Cao, G., et al. (2003). Tumor-derived vascular endothelial growth factor up-regulates angiopoietin-2 in host endothelium and destabilizes host vasculature, supporting angiogenesis in ovarian cancer. Cancer Research, 63, 3403–412.

    PubMed  CAS  Google Scholar 

  94. Dome, B., Paku, S., Somlai, B., & Timar, J. (2002). Vascularization of cutaneous melanoma involves vessel co-option and has clinical significance. Journal of Pathology, 197, 355–62.

    PubMed  Google Scholar 

  95. Kim, E. S., Serur, A., Huang, J., Manley, C. A., McCrudden, K. W., Frischer, J. S., et al. (2002). Potent VEGF blockade causes regression of coopted vessels in a model of neuroblastoma. Proceedings of the National Academy of Sciences of the United States of America, 99, 11399–1404.

    PubMed  CAS  Google Scholar 

  96. Kunkel, P., Ulbricht, U., Bohlen, P., Brockmann, M. A., Fillbrandt, R., Stavrou, D., et al. (2001). Inhibition of glioma angiogenesis and growth in vivo by systemic treatment with a monoclonal antibody against vascular endothelial growth factor receptor-2. Cancer Research, 61, 6624–628.

    PubMed  CAS  Google Scholar 

  97. Maniotis, A. J., Folberg, R., Hess, A., Seftor, E. A., Gardner, L. M., Pe’er, J., et al. (1999). Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. American Journal of Pathology, 155, 739–52.

    PubMed  CAS  Google Scholar 

  98. Folberg, R., Rummelt, V., Parys-Van Ginderdeuren, R., Hwang, T., Woolson, R. F., Pe’er, J., et al. (1993). The prognostic value of tumor blood vessel morphology in primary uveal melanoma. Ophthalmology, 100, 1389–398.

    PubMed  CAS  Google Scholar 

  99. Hendrix, M. J., Seftor, E. A., Hess, A. R., & Seftor, R. E. (2003). Vasculogenic mimicry and tumour-cell plasticity: Lessons from melanoma. Nature Reviews Cancer, 3, 411–21.

    PubMed  CAS  Google Scholar 

  100. Seftor, R. E., Seftor, E. A., Koshikawa, N., Meltzer, P. S., Gardner, L. M., Bilban, M., et al. (2001). Cooperative interactions of laminin 5 gamma2 chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Research, 61, 6322–327.

    PubMed  CAS  Google Scholar 

  101. Hendrix, M. J., Seftor, E. A., Meltzer, P. S., Gardner, L. M., Hess, A. R., Kirschmann, D. A., et al. (2001). Expression and functional significance of VE-cadherin in aggressive human melanoma cells: Role in vasculogenic mimicry. Proceedings of the National Academy of Sciences of the United States of America, 98, 8018–023.

    PubMed  CAS  Google Scholar 

  102. Hess, A. R., Postovit, L. M., Margaryan, N. V., Seftor, E. A., Schneider, G. B., Seftor, R. E., et al. (2005). Focal adhesion kinase promotes the aggressive melanoma phenotype. Cancer Research, 65, 9851–860.

    PubMed  CAS  Google Scholar 

  103. Basu, G. D., Liang, W. S., Stephan, D. A., Wegener, L. T., Conley, C. R., Pockaj, B. A., et al. (2006). A novel role for cyclooxygenase-2 in regulating vascular channel formation by human breast cancer cells. Breast Cancer Research, 8, R69.

    PubMed  Google Scholar 

  104. Rothhammer, T., Bataille, F., Spruss, T., Eissner, G., & Bosserhoff, A. K. (2007). Functional implication of BMP4 expression on angiogenesis in malignant melanoma. Oncogene, 26, 4158–170.

    PubMed  CAS  Google Scholar 

  105. Xi, Y., Nakajima, G., Hamil, T., Fodstad, O., Riker, A., & Ju, J. (2006). Association of insulin-like growth factor binding protein-3 expression with melanoma progression. Molecular Cancer Therapeutics, 5, 3078–084.

    PubMed  CAS  Google Scholar 

  106. Topczewska, J. M., Postovit, L. M., Margaryan, N. V., Sam, A., Hess, A. R., Wheaton, W. W., et al. (2006). Embryonic and tumorigenic pathways converge via Nodal signaling: Role in melanoma aggressiveness. Nature Medicine, 12, 925–32.

    PubMed  CAS  Google Scholar 

  107. Seftor, E. A., Brown, K. M., Chin, L., Kirschmann, D. A., Wheaton, W. W., Protopopov, A., et al. (2005). Epigenetic transdifferentiation of normal melanocytes by a metastatic melanoma microenvironment. Cancer Research, 65, 10164–0169.

    PubMed  CAS  Google Scholar 

  108. Rofstad, E. K., Rasmussen, H., Galappathi, K., Mathiesen, B., Nilsen, K., & Graff, B. A. (2002). Hypoxia promotes lymph node metastasis in human melanoma xenografts by up-regulating the urokinase-type plasminogen activator receptor. Cancer Research, 62, 1847–853.

    PubMed  CAS  Google Scholar 

  109. Bedogni, B., Welford, S. M., Cassarino, D. S., Nickoloff, B. J., Giaccia, A. J., & Powell, M. B. (2005). The hypoxic microenvironment of the skin contributes to Akt-mediated melanocyte transformation. Cancer Cell, 8, 443–54.

    PubMed  CAS  Google Scholar 

  110. Rybak, S. M., Sanovich, E., Hollingshead, M. G., Borgel, S. D., Newton, D. L., Melillo, G., et al. (2003). “Vasocrine–formation of tumor cell-lined vascular spaces: Implications for rational design of antiangiogenic therapies. Cancer Research, 63, 2812–819.

    PubMed  CAS  Google Scholar 

  111. van der Schaft, D. W., Hillen, F., Pauwels, P., Kirschmann, D. A., Castermans, K., Egbrink, M. G., et al. (2005). Tumor cell plasticity in Ewing sarcoma, an alternative circulatory system stimulated by hypoxia. Cancer Research, 65, 11520–1528.

    PubMed  Google Scholar 

  112. Sun, B., Zhang, D., Zhang, S., Zhang, W., Guo, H., & Zhao, X. (2007). Hypoxia influences vasculogenic mimicry channel formation and tumor invasion-related protein expression in melanoma. Cancer Letter, 249, 188–97.

    CAS  Google Scholar 

  113. Clarijs, R., Otte-Holler, I., Ruiter, D. J., & de Waal, R. M. (2002). Presence of a fluid-conducting meshwork in xenografted cutaneous and primary human uveal melanoma. Investigative Ophthalmology and Visual Science, 43, 912–18.

    PubMed  Google Scholar 

  114. Shirakawa, K., Kobayashi, H., Heike, Y., Kawamoto, S., Brechbiel, M. W., Kasumi, F., et al. (2002). Hemodynamics in vasculogenic mimicry and angiogenesis of inflammatory breast cancer xenograft. Cancer Research, 62, 560–66.

    PubMed  CAS  Google Scholar 

  115. Ruf, W., Seftor, E. A., Petrovan, R. J., Weiss, R. M., Gruman, L. M., Margaryan, N. V., et al. (2003). Differential role of tissue factor pathway inhibitors 1 and 2 in melanoma vasculogenic mimicry. Cancer Research, 63, 5381–389.

    PubMed  CAS  Google Scholar 

  116. Frenkel, S., Barzel, I., Levy, J., Lin, A. Y., Bartsch, D. U., Majumdar, D., Folberg, R., & Pe’er, J. (2007). Demonstrating circulation in vasculogenic mimicry patterns of uveal melanoma by confocal indocyanine green angiography. Eye, in press, DOI 10.1038/sj.eye.6702783.

  117. Thies, A., Mangold, U., Moll, I., & Schumacher, U. (2001). PAS-positive loops and networks as a prognostic indicator in cutaneous malignant melanoma. Journal of Pathology, 195, 537–42.

    PubMed  CAS  Google Scholar 

  118. Lee, Y. J., Nagai, N., Siar, C. H., Nakano, K., Nagatsuka, H., Tsujigiwa, H., et al. (2002). Angioarchitecture of primary oral malignant melanomas. Journal of Histochemistry and Cytochemistry, 50, 1555–562.

    PubMed  CAS  Google Scholar 

  119. Sharma, N., Seftor, R. E., Seftor, E. A., Gruman, L. M., Heidger, P. M., Jr., Cohen, M. B., et al. (2002). Prostatic tumor cell plasticity involves cooperative interactions of distinct phenotypic subpopulations: Role in vasculogenic mimicry. Prostate, 50, 189–01.

    PubMed  Google Scholar 

  120. Sood, A. K., Seftor, E. A., Fletcher, M. S., Gardner, L. M., Heidger, P. M., Buller, R. E., et al. (2001). Molecular determinants of ovarian cancer plasticity. American Journal of Pathology, 158, 1279–288.

    PubMed  CAS  Google Scholar 

  121. Sun, B., Zhang, S., Zhang, D., Du, J., Guo, H., Zhao, X., et al. (2006). Vasculogenic mimicry is associated with high tumor grade, invasion and metastasis, and short survival in patients with hepatocellular carcinoma. Oncology Reports, 16, 693–98.

    PubMed  CAS  Google Scholar 

  122. Fujimoto, A., Onodera, H., Mori, A., Nagayama, S., Yonenaga, Y., & Tachibana, T. (2006). Tumour plasticity and extravascular circulation in ECV304 human bladder carcinoma cells. Anticancer Research, 26, 59–9.

    PubMed  Google Scholar 

  123. Sun, B., Zhang, S., Zhao, X., Zhang, W., & Hao, X. (2004). Vasculogenic mimicry is associated with poor survival in patients with mesothelial sarcomas and alveolar rhabdomyosarcomas. International Journal of Oncology, 25, 1609–614.

    PubMed  Google Scholar 

  124. Cai, X. S., Jia, Y. W., Mei, J., & Tang, R. Y. (2004). Tumor blood vessels formation in osteosarcoma: Vasculogenesis mimicry. Chinese Medical Journal (Engl), 117, 94–8.

    Google Scholar 

  125. Yue, W. Y. & Chen, Z. P. (2005). Does vasculogenic mimicry exist in astrocytoma? Journal of Histochemistry and Cytochemistry, 53, 997–002.

    PubMed  CAS  Google Scholar 

  126. Favier, J., Plouin, P. F., Corvol, P., & Gasc, J. M. (2002). Angiogenesis and vascular architecture in pheochromocytomas: Distinctive traits in malignant tumors. American Journal of Pathology, 161, 1235–246.

    PubMed  CAS  Google Scholar 

  127. Seftor, R. E., Seftor, E. A., Kirschmann, D. A., & Hendrix, M. J. (2002). Targeting the tumor microenvironment with chemically modified tetracyclines: inhibition of laminin 5 gamma2 chain promigratory fragments and vasculogenic mimicry. Molecular Cancer Therapeutics, 1, 1173–179.

    PubMed  CAS  Google Scholar 

  128. Rudek, M. A., Horne, M., Figg, W. D., Dahut, W., Dyer, V., Pluda, J. M., et al. (2001). Reversible sideroblastic anemia associated with the tetracycline analogue COL-3. American Journal of Hematology, 67, 51–3.

    PubMed  CAS  Google Scholar 

  129. Ghate, J. V., Turner, M. L., Rudek, M. A., Figg, W. D., Dahut, W., Dyer, V., et al. (2001). Drug-induced lupus associated with COL-3: Report of 3 cases. Archives of Dermatology, 137, 471–74.

    PubMed  CAS  Google Scholar 

  130. van der Schaft, D. W., Seftor, R. E., Seftor, E. A., Hess, A. R., Gruman, L. M., Kirschmann, D. A., et al. (2004). Effects of angiogenesis inhibitors on vascular network formation by human endothelial and melanoma cells. Journal of the National Cancer Institute, 96, 1473–477.

    Article  PubMed  Google Scholar 

  131. Pepper, M. S., & Skobe, M. (2003). Lymphatic endothelium: Morphological, molecular and functional properties. Journal of Cell Biology, 163, 209–13.

    PubMed  CAS  Google Scholar 

  132. Perou, C. M., Sorlie, T., Eisen, M. B., van de Rijn, M., Jeffrey, S. S., Rees, C. A., et al. (2000). Molecular portraits of human breast tumours. Nature, 406, 747–52.

    PubMed  CAS  Google Scholar 

  133. Podgrabinska, S., Braun, P., Velasco, P., Kloos, B., Pepper, M. S., & Skobe, M. (2002). Molecular characterization of lymphatic endothelial cells. Proceedings of the National Academy of Sciences of the United States of America, 99, 16069–6074.

    PubMed  CAS  Google Scholar 

  134. Kaipainen, A., Korhonen, J., Mustonen, T., van Hinsbergh, V. W., Fang, G. H., Dumont, D., et al. (1995). Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proceedings of the National Academy of Sciences of the United States of America, 92, 3566–570.

    PubMed  CAS  Google Scholar 

  135. Banerji, S., Ni, J., Wang, S. X., Clasper, S., Su, J., Tammi, R., et al. (1999). LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for hyaluronan. Journal of Cell Biology, 144, 789–01.

    PubMed  CAS  Google Scholar 

  136. Breiteneder-Geleff, S., Soleiman, A., Kowalski, H., Horvat, R., Amann, G., Kriehuber, E., et al. (1999). Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: Podoplanin as a specific marker for lymphatic endothelium. American Journal of Pathology, 154, 385–94.

    PubMed  CAS  Google Scholar 

  137. Oliver, G., & Detmar, M. (2002). The rediscovery of the lymphatic system: Old and new insights into the development and biological function of the lymphatic vasculature. Genes & Development, 16, 773–83.

    CAS  Google Scholar 

  138. Joukov, V., Kumar, V., Sorsa, T., Arighi, E., Weich, H., Saksela, O., et al. (1998). A recombinant mutant vascular endothelial growth factor-C that has lost vascular endothelial growth factor receptor-2 binding, activation, and vascular permeability activities. Journal of Biological Chemistry, 273, 6599–602.

    PubMed  CAS  Google Scholar 

  139. Baldwin, M. E., Halford, M. M., Roufail, S., Williams, R. A., Hibbs, M. L., Grail, D., et al. (2005). Vascular endothelial growth factor D is dispensable for development of the lymphatic system. Molular and Cellular Biology, 25, 2441–449.

    PubMed  CAS  Google Scholar 

  140. Karkkainen, M. J., Haiko, P., Sainio, K., Partanen, J., Taipale, J., Petrova, T. V., et al. (2004). Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nature Immunology, 5, 74–0.

    PubMed  CAS  Google Scholar 

  141. Makinen, T., Veikkola, T., Mustjoki, S., Karpanen, T., Catimel, B., Nice, E. C., et al. (2001). Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C/D receptor VEGFR-3. EMBO Journal, 20, 4762–773.

    PubMed  CAS  Google Scholar 

  142. Skobe, M., Hawighorst, T., Jackson, D. G., Prevo, R., Janes, L., Velasco, P., et al. (2001). Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nature Medicine, 7, 192–98.

    PubMed  CAS  Google Scholar 

  143. Stacker, S. A., Caesar, C., Baldwin, M. E., Thornton, G. E., Williams, R. A., Prevo, R., et al. (2001). VEGF-D promotes the metastatic spread of tumor cells via the lymphatics. Nature Medicine, 7, 186–91.

    PubMed  CAS  Google Scholar 

  144. Nagy, J. A., Vasile, E., Feng, D., Sundberg, C., Brown, L. F., Detmar, M. J., et al. (2002). Vascular permeability factor/vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. Journal of Experimental Medicine, 196, 1497–506.

    PubMed  CAS  Google Scholar 

  145. Chang, L. K., Garcia-Cardena, G., Farnebo, F., Fannon, M., Chen, E. J., Butterfield, C., et al. (2004). Dose-dependent response of FGF-2 for lymphangiogenesis. Proceedings of the National Academy of Sciences of the United States of America, 101, 11658–1663.

    PubMed  CAS  Google Scholar 

  146. Cao, R., Bjorndahl, M. A., Religa, P., Clasper, S., Garvin, S., Galter, D., et al. (2004). PDGF-BB induces intratumoral lymphangiogenesis and promotes lymphatic metastasis. Cancer Cell, 6, 333–45.

    PubMed  CAS  Google Scholar 

  147. Anan, K., Morisaki, T., Katano, M., Ikubo, A., Kitsuki, H., Uchiyama, A., et al. (1996). Vascular endothelial growth factor and platelet-derived growth factor are potential angiogenic and metastatic factors in human breast cancer. Surgery, 119, 333–39.

    PubMed  CAS  Google Scholar 

  148. Gale, N. W., Thurston, G., Hackett, S. F., Renard, R., Wang, Q., McClain, J., et al. (2002). Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1. Dev Cell, 3, 411–23.

    PubMed  CAS  Google Scholar 

  149. Morisada, T., Oike, Y., Yamada, Y., Urano, T., Akao, M., Kubota, Y., et al. (2005). Angiopoietin-1 promotes LYVE-1-positive lymphatic vessel formation. Blood, 105, 4649–656.

    PubMed  CAS  Google Scholar 

  150. Yuan, L., Moyon, D., Pardanaud, L., Breant, C., Karkkainen, M. J., Alitalo, K., et al. (2002). Abnormal lymphatic vessel development in neuropilin 2 mutant mice. Development, 129, 4797–806.

    PubMed  CAS  Google Scholar 

  151. Kajiya, K., Hirakawa, S., Ma, B., Drinnenberg, I., & Detmar, M. (2005). Hepatocyte growth factor promotes lymphatic vessel formation and function. EMBO Journal, 24, 2885–895.

    PubMed  CAS  Google Scholar 

  152. Bjorndahl, M., Cao, R., Nissen, L. J., Clasper, S., Johnson, L. A., Xue, Y., et al. (2005). Insulin-like growth factors 1 and 2 induce lymphangiogenesis in vivo. Proceedings of the National Academy of Sciences of the United States of America, 102, 15593–5598.

    PubMed  Google Scholar 

  153. Saharinen, P., Tammela, T., Karkkainen, M. J., & Alitalo, K. (2004). Lymphatic vasculature: development, molecular regulation and role in tumor metastasis and inflammation. Trends in Immunology, 25, 387–95.

    PubMed  CAS  Google Scholar 

  154. Ji, R. C. (2006). Lymphatic endothelial cells, tumor lymphangiogenesis and metastasis: New insights into intratumoral and peritumoral lymphatics. Cancer and Metastasis Reviews, 25, 677–94.

    PubMed  Google Scholar 

  155. Achen, M. G., Mann, G. B., & Stacker, S. A. (2006). Targeting lymphangiogenesis to prevent tumour metastasis. British Journal of Cancer, 94, 1355–360.

    PubMed  CAS  Google Scholar 

  156. Stacker, S. A., Achen, M. G., Jussila, L., Baldwin, M. E., & Alitalo, K. (2002). Lymphangiogenesis and cancer metastasis. Nature Reviews Cancer, 2, 573–83.

    CAS  Google Scholar 

  157. Schoppmann, S. F., Bayer, G., Aumayr, K., Taucher, S., Geleff, S., Rudas, M., et al. (2004). Prognostic value of lymphangiogenesis and lymphovascular invasion in invasive breast cancer. Annals of Surgery, 240, 306–12.

    PubMed  Google Scholar 

  158. Lee, A. H., Pinder, S. E., Macmillan, R. D., Mitchell, M., Ellis, I. O., Elston, C. W., et al. (2006). Prognostic value of lymphovascular invasion in women with lymph node negative invasive breast carcinoma. European Journal of Cancer, 42, 357–62.

    PubMed  CAS  Google Scholar 

  159. van der Schaft, D. W., Pauwels, P., Hulsmans, S., Zimmermann, M., van de Poll-Franse, L. V., & Griffioen, A. W. (2007). Absence of lymphangiogenesis in ductal breast cancer at the primary tumor site. Cancer Letter, 254, 128–36.

    Google Scholar 

  160. Arigami, T., Natsugoe, S., Uenosono, Y., Arima, H., Mataki, Y., Ehi, K., et al. (2005). Lymphatic invasion using D2-40 monoclonal antibody and its relationship to lymph node micrometastasis in pN0 gastric cancer. British Journal of Cancer, 93, 688–93.

    PubMed  CAS  Google Scholar 

  161. Lotan, Y., Gupta, A., Shariat, S. F., Palapattu, G. S., Vazina, A., Karakiewicz, P. I., et al. (2005). Lymphovascular invasion is independently associated with overall survival, cause-specific survival, and local and distant recurrence in patients with negative lymph nodes at radical cystectomy. Journal of Clinical Oncology, 23, 6533–539.

    PubMed  Google Scholar 

  162. Karpanen, T., Egeblad, M., Karkkainen, M. J., Kubo, H., Yla-Herttuala, S., Jaattela, M., et al. (2001). Vascular endothelial growth factor C promotes tumor lymphangiogenesis and intralymphatic tumor growth. Cancer Research, 61, 1786–790.

    PubMed  CAS  Google Scholar 

  163. Szuba, A., Skobe, M., Karkkainen, M. J., Shin, W. S., Beynet, D. P., Rockson, N. B., et al. (2002). Therapeutic lymphangiogenesis with human recombinant VEGF-C. FASEB Journal, 16, 1985–987.

    PubMed  CAS  Google Scholar 

  164. Zhang, L., Giraudo, E., Hoffman, J. A., Hanahan, D., & Ruoslahti, E. (2006). Lymphatic zip codes in premalignant lesions and tumors. Cancer Research, 66, 5696–706.

    PubMed  CAS  Google Scholar 

  165. Jain, R. K. (2005). Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science, 307, 58–2.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Arjan W. Griffioen.

About this article

Cite this article

Hillen, F., Griffioen, A.W. Tumour vascularization: sprouting angiogenesis and beyond. Cancer Metastasis Rev 26, 489–502 (2007). https://doi.org/10.1007/s10555-007-9094-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10555-007-9094-7

Keywords

Navigation