Skip to main content

Advertisement

Log in

Oncogenes and angiogenesis: a way to personalize anti-angiogenic therapy?

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

The acquisition of oncogenic mutations and promotion of angiogenesis are key hallmarks of cancer. These features are often thought of as separate events in tumor progression and the two fields of research have frequently been considered as independent. However, as we highlight in this review, activated oncogenes and deregulated angiogenesis are tightly associated, as mutations in cancer cells can lead to perturbation of the pro- and anti-angiogenic balance thereby causing aberrant angiogenesis. We propose that normalization of the vascular network by targeting oncogenes in the tumor cells might lead to more efficient and sustained therapeutic effects compared to therapies targeting tumor vessels. We discuss how pharmacological inhibition of oncogenes in tumor cells restores a functional vasculature by bystander anti-angiogenic effect. As genetic alterations are tumor-specific, targeted therapy, which potentially blocks the angiogenic program activated by individual oncogenes may lead to personalized anti-angiogenic therapy.

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. Risau W (1997) Mechanisms of angiogenesis. Nature 386(6626):671–674. doi:10.1038/386671a0

    CAS  PubMed  Google Scholar 

  2. Carmeliet P, Jain RK (2000) Angiogenesis in cancer and other diseases. Nature 407(6801):249–257

    CAS  PubMed  Google Scholar 

  3. Folkman J (1971) Tumor angiogenesis: therapeutic implications. N Engl J Med 285(21):1182–1186. doi:10.1056/NEJM197111182852108

    CAS  PubMed  Google Scholar 

  4. Folkman J (2006) Angiogenesis. Annu Rev Med 57:1–18. doi:10.1146/annurev.med.57.121304.131306

    CAS  PubMed  Google Scholar 

  5. Bergers G, Benjamin LE (2003) Tumorigenesis and the angiogenic switch. Nat Rev Cancer 3(6):401–410

    CAS  PubMed  Google Scholar 

  6. Morikawa S, Baluk P, Kaidoh T, Haskell A, Jain RK, McDonald DM (2002) Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am J Pathol 160(3):985–1000. doi:10.1016/S0002-9440(10)64920-6

    PubMed  Google Scholar 

  7. Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G, Roberge S, Jain RK, McDonald DM (2000) Openings between defective endothelial cells explain tumor vessel leakiness. Am J Pathol 156(4):1363–1380. doi:10.1016/S0002-9440(10)65006-7

    CAS  PubMed  Google Scholar 

  8. Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF (1983) Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 219(4587):983–985

    CAS  PubMed  Google Scholar 

  9. Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N (1989) Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246(4935):1306–1309

    CAS  PubMed  Google Scholar 

  10. Keck PJ, Hauser SD, Krivi G, Sanzo K, Warren T, Feder J, Connolly DT (1989) Vascular permeability factor, an endothelial cell mitogen related to PDGF. Science 246(4935):1309–1312

    CAS  PubMed  Google Scholar 

  11. Ferrara N, Hillan KJ, Gerber HP, Novotny W (2004) Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 3(5):391–400. doi:10.1038/nrd1381

    CAS  PubMed  Google Scholar 

  12. Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M, Cao Y, Shujath J, Gawlak S, Eveleigh D, Rowley B, Liu L, Adnane L, Lynch M, Auclair D, Taylor I, Gedrich R, Voznesensky A, Riedl B, Post LE, Bollag G, Trail PA (2004) BAY 43–9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res 64(19):7099–7109. doi:10.1158/0008-5472.CAN-04-1443

    CAS  PubMed  Google Scholar 

  13. Strumberg D, Richly H, Hilger RA, Schleucher N, Korfee S, Tewes M, Faghih M, Brendel E, Voliotis D, Haase CG, Schwartz B, Awada A, Voigtmann R, Scheulen ME, Seeber S (2005) Phase I clinical and pharmacokinetic study of the Novel Raf kinase and vascular endothelial growth factor receptor inhibitor BAY 43–9006 in patients with advanced refractory solid tumors. J Clin Oncol 23(5):965–972. doi:10.1200/JCO.2005.06.124

    CAS  PubMed  Google Scholar 

  14. Motzer RJ, Michaelson MD, Redman BG, Hudes GR, Wilding G, Figlin RA, Ginsberg MS, Kim ST, Baum CM, DePrimo SE, Li JZ, Bello CL, Theuer CP, George DJ, Rini BI (2006) Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma. J Clin Oncol 24(1):16–24. doi:10.1200/JCO.2005.02.2574

    CAS  PubMed  Google Scholar 

  15. Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, Ferrara N (1993) Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362(6423):841–844. doi:10.1038/362841a0

    CAS  PubMed  Google Scholar 

  16. Asano M, Yukita A, Matsumoto T, Kondo S, Suzuki H (1995) Inhibition of tumor growth and metastasis by an immunoneutralizing monoclonal antibody to human vascular endothelial growth factor/vascular permeability factor121. Cancer Res 55(22):5296–5301

    CAS  PubMed  Google Scholar 

  17. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350(23):2335–2342. doi:10.1056/NEJMoa032691

    CAS  PubMed  Google Scholar 

  18. Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Archer L, Atkins JN, Picus J, Czaykowski P, Dutcher J, Small EJ (2010) Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal cell carcinoma: final results of CALGB 90206. J Clin Oncol 28(13):2137–2143. doi:10.1200/JCO.2009.26.5561

    CAS  PubMed  Google Scholar 

  19. Cloughesy T (2010) FDA accelerated approval benefits glioblastoma. Lancet Oncol 11(12):1120. doi:10.1016/S1470-2045(10)70269-2

    PubMed  Google Scholar 

  20. Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, Shenkier T, Cella D, Davidson NE (2007) Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 357(26):2666–2676. doi:10.1056/NEJMoa072113

    CAS  PubMed  Google Scholar 

  21. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, Lilenbaum R, Johnson DH (2006) Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 355(24):2542–2550. doi:10.1056/NEJMoa061884

    CAS  PubMed  Google Scholar 

  22. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F, Demkow T, Hutson TE, Gore M, Freeman S, Schwartz B, Shan M, Simantov R, Bukowski RM (2007) Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 356(2):125–134. doi:10.1056/NEJMoa060655

    CAS  PubMed  Google Scholar 

  23. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, Schwartz M, Porta C, Zeuzem S, Bolondi L, Greten TF, Galle PR, Seitz JF, Borbath I, Haussinger D, Giannaris T, Shan M, Moscovici M, Voliotis D, Bruix J (2008) Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 359(4):378–390. doi:10.1056/NEJMoa0708857

    CAS  PubMed  Google Scholar 

  24. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, Negrier S, Szczylik C, Pili R, Bjarnason GA, Garcia-del-Muro X, Sosman JA, Solska E, Wilding G, Thompson JA, Kim ST, Chen I, Huang X, Figlin RA (2009) Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol 27(22):3584–3590. doi:10.1200/JCO.2008.20.1293

    CAS  PubMed  Google Scholar 

  25. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, Zarba JJ, Chen M, McCann L, Pandite L, Roychowdhury DF, Hawkins RE (2010) Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 28(6):1061–1068. doi:10.1200/JCO.2009.23.9764

    CAS  PubMed  Google Scholar 

  26. Kerbel RS (2008) Tumor angiogenesis. N Engl J Med 358(19):2039–2049. doi:10.1056/NEJMra0706596

    CAS  PubMed  Google Scholar 

  27. Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 8(8):592–603. doi:10.1038/nrc2442

    CAS  PubMed  Google Scholar 

  28. Boneberg EM, Legler DF, Hoefer MM, Ohlschlegel C, Steininger H, Fuzesi L, Beer GM, Dupont-Lampert V, Otto F, Senn HJ, Furstenberger G (2009) Angiogenesis and lymphangiogenesis are downregulated in primary breast cancer. Br J Cancer 101(4):605–614. doi:10.1038/sj.bjc.6605219

    CAS  PubMed  Google Scholar 

  29. Yu JL, Rak JW, Carmeliet P, Nagy A, Kerbel RS, Coomber BL (2001) Heterogeneous vascular dependence of tumor cell populations. Am J Pathol 158(4):1325–1334. doi:10.1016/S0002-9440(10)64083-7

    CAS  PubMed  Google Scholar 

  30. Leenders WP, Kusters B, de Waal RM (2002) Vessel co-option: how tumors obtain blood supply in the absence of sprouting angiogenesis. Endothelium 9(2):83–87

    PubMed  Google Scholar 

  31. Hendrix MJ, Seftor RE, Seftor EA, Gruman LM, Lee LM, Nickoloff BJ, Miele L, Sheriff DD, Schatteman GC (2002) Transendothelial function of human metastatic melanoma cells: role of the microenvironment in cell-fate determination. Cancer Res 62(3):665–668

    CAS  PubMed  Google Scholar 

  32. Ricci-Vitiani L, Pallini R, Biffoni M, Todaro M, Invernici G, Cenci T, Maira G, Parati EA, Stassi G, Larocca LM, De Maria R (2010) Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 468(7325):824–828. doi:10.1038/nature09557

    CAS  PubMed  Google Scholar 

  33. Wang R, Chadalavada K, Wilshire J, Kowalik U, Hovinga KE, Geber A, Fligelman B, Leversha M, Brennan C, Tabar V (2010) Glioblastoma stem-like cells give rise to tumour endothelium. Nature 468(7325):829–833. doi:10.1038/nature09624

    CAS  PubMed  Google Scholar 

  34. Peters BA, Diaz LA, Polyak K, Meszler L, Romans K, Guinan EC, Antin JH, Myerson D, Hamilton SR, Vogelstein B, Kinzler KW, Lengauer C (2005) Contribution of bone marrow-derived endothelial cells to human tumor vasculature. Nat Med 11(3):261–262. doi:10.1038/nm1200

    CAS  PubMed  Google Scholar 

  35. Leenders WP, Kusters B, Verrijp K, Maass C, Wesseling P, Heerschap A, Ruiter D, Ryan A, de Waal R (2004) Antiangiogenic therapy of cerebral melanoma metastases results in sustained tumor progression via vessel co-option. Clin Cancer Res 10(18 Pt 1):6222–6230. doi:10.1158/1078-0432.CCR-04-0823

    CAS  PubMed  Google Scholar 

  36. Rubenstein JL, Kim J, Ozawa T, Zhang M, Westphal M, Deen DF, Shuman MA (2000) Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia 2(4):306–314

    CAS  PubMed  Google Scholar 

  37. Relf M, LeJeune S, Scott PA, Fox S, Smith K, Leek R, Moghaddam A, Whitehouse R, Bicknell R, Harris AL (1997) Expression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor beta-1, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesis. Cancer Res 57(5):963–969

    CAS  PubMed  Google Scholar 

  38. Pollard JW (2004) Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer 4(1):71–78. doi:10.1038/nrc1256

    CAS  PubMed  Google Scholar 

  39. Orimo A, Weinberg RA (2006) Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle 5(15):1597–1601. doi:3112

    CAS  PubMed  Google Scholar 

  40. Shojaei F, Ferrara N (2008) Refractoriness to antivascular endothelial growth factor treatment: role of myeloid cells. Cancer Res 68(14):5501–5504. doi:10.1158/0008-5472.CAN-08-0925

    CAS  PubMed  Google Scholar 

  41. Casanovas O, Hicklin DJ, Bergers G, Hanahan D (2005) Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8(4):299–309. doi:10.1016/j.ccr.2005.09.005

    CAS  PubMed  Google Scholar 

  42. Batchelor TT, Sorensen AG, di Tomaso E, Zhang WT, Duda DG, Cohen KS, Kozak KR, Cahill DP, Chen PJ, Zhu M, Ancukiewicz M, Mrugala MM, Plotkin S, Drappatz J, Louis DN, Ivy P, Scadden DT, Benner T, Loeffler JS, Wen PY, Jain RK (2007) AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 11(1):83–95. doi:10.1016/j.ccr.2006.11.021

    CAS  PubMed  Google Scholar 

  43. Crawford Y, Kasman I, Yu L, Zhong C, Wu X, Modrusan Z, Kaminker J, Ferrara N (2009) PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Cancer Cell 15(1):21–34. doi:10.1016/j.ccr.2008.12.004

    CAS  PubMed  Google Scholar 

  44. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA (2005) Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121(3):335–348. doi:10.1016/j.cell.2005.02.034

    CAS  PubMed  Google Scholar 

  45. Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, Fuh G, Gerber HP, Ferrara N (2007) Tumor refractoriness to anti-VEGF treatment is mediated by CD11b + Gr1 + myeloid cells. Nat Biotechnol 25(8):911–920. doi:10.1038/nbt1323

    CAS  PubMed  Google Scholar 

  46. Shojaei F, Wu X, Zhong C, Yu L, Liang XH, Yao J, Blanchard D, Bais C, Peale FV, van Bruggen N, Ho C, Ross J, Tan M, Carano RA, Meng YG, Ferrara N (2007) Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature 450(7171):825–831. doi:10.1038/nature06348

    CAS  PubMed  Google Scholar 

  47. Shojaei F, Lee JH, Simmons BH, Wong A, Esparza CO, Plumlee PA, Feng J, Stewart AE, Hu-Lowe DD, Christensen JG (2010) HGF/c-Met acts as an alternative angiogenic pathway in sunitinib-resistant tumors. Cancer Res 70(24):10090–10100. doi:10.1158/0008-5472.CAN-10-0489

    CAS  PubMed  Google Scholar 

  48. Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS (2009) Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15(3):232–239. doi:10.1016/j.ccr.2009.01.021

    CAS  PubMed  Google Scholar 

  49. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, Inoue M, Bergers G, Hanahan D, Casanovas O (2009) Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15(3):220–231. doi:10.1016/j.ccr.2009.01.027

    CAS  PubMed  Google Scholar 

  50. Loges S, Mazzone M, Hohensinner P, Carmeliet P (2009) Silencing or fueling metastasis with VEGF inhibitors: antiangiogenesis revisited. Cancer Cell 15(3):167–170. doi:10.1016/j.ccr.2009.02.007

    CAS  PubMed  Google Scholar 

  51. Miles D, Harbeck N, Escudier B, Hurwitz H, Saltz L, Van Cutsem E, Cassidy J, Mueller B, Sirzen F (2011) Disease course patterns after discontinuation of bevacizumab: pooled analysis of randomized phase III trials. J Clin Oncol 29(1):83–88. doi:10.1200/JCO.2010.30.2794

    CAS  PubMed  Google Scholar 

  52. Zuniga RM, Torcuator R, Jain R, Anderson J, Doyle T, Schultz L, Mikkelsen T (2010) Rebound tumour progression after the cessation of bevacizumab therapy in patients with recurrent high-grade glioma. J Neurooncol 99(2):237–242. doi:10.1007/s11060-010-0121-0

    CAS  PubMed  Google Scholar 

  53. Verhoeff JJ, van Tellingen O, Claes A, Stalpers LJ, van Linde ME, Richel DJ, Leenders WP, van Furth WR (2009) Concerns about anti-angiogenic treatment in patients with glioblastoma multiforme. BMC Cancer 9:444. doi:10.1186/1471-2407-9-444

    PubMed  Google Scholar 

  54. Johannsen M, Florcken A, Bex A, Roigas J, Cosentino M, Ficarra V, Kloeters C, Rief M, Rogalla P, Miller K, Grunwald V (2009) Can tyrosine kinase inhibitors be discontinued in patients with metastatic renal cell carcinoma and a complete response to treatment? A multicentre, retrospective analysis. Eur Urol 55(6):1430–1438. doi:10.1016/j.eururo.2008.10.021

    CAS  PubMed  Google Scholar 

  55. Soeda A, Park M, Lee D, Mintz A, Androutsellis-Theotokis A, McKay RD, Engh J, Iwama T, Kunisada T, Kassam AB, Pollack IF, Park DM (2009) Hypoxia promotes expansion of the CD133-positive glioma stem cells through activation of HIF-1alpha. Oncogene 28(45):3949–3959. doi:10.1038/onc.2009.252

    CAS  PubMed  Google Scholar 

  56. Heddleston JM, Li Z, McLendon RE, Hjelmeland AB, Rich JN (2009) The hypoxic microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a cancer stem cell phenotype. Cell Cycle 8(20):3274–3284. doi:9701

    CAS  PubMed  Google Scholar 

  57. Conley SJ, Gheordunescu E, Kakarala P, Newman B, Korkaya H, Heath AN, Clouthier SG, Wicha MS (2012) Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia. Proc Natl Acad Sci USA 109(8):2784–2789. doi:10.1073/pnas.1018866109

    CAS  PubMed  Google Scholar 

  58. Visvader JE, Lindeman GJ (2008) Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8(10):755–768. doi:10.1038/nrc2499

    CAS  PubMed  Google Scholar 

  59. Yu Y, Ramena G, Elble RC (2012) The role of cancer stem cells in relapse of solid tumors. Front Biosci (Elite Ed) 4:1528–1541. doi:478

    Google Scholar 

  60. Dallas NA, Xia L, Fan F, Gray MJ, Gaur P, Van Buren G 2nd, Samuel S, Kim MP, Lim SJ, Ellis LM (2009) Chemoresistant colorectal cancer cells, the cancer stem cell phenotype, and increased sensitivity to insulin-like growth factor-I receptor inhibition. Cancer Res 69(5):1951–1957. doi:10.1158/0008-5472.CAN-08-2023

    CAS  PubMed  Google Scholar 

  61. Baluk P, Morikawa S, Haskell A, Mancuso M, McDonald DM (2003) Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am J Pathol 163(5):1801–1815. doi:10.1016/S0002-9440(10)63540-7

    PubMed  Google Scholar 

  62. De Bock K, Cauwenberghs S, Carmeliet P (2011) Vessel abnormalization: another hallmark of cancer? Molecular mechanisms and therapeutic implications. Curr Opin Genet Dev 21(1):73–79. doi:10.1016/j.gde.2010.10.008

    PubMed  Google Scholar 

  63. Jain RK (2005) Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307(5706):58–62

    CAS  PubMed  Google Scholar 

  64. Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, Jain RK (2011) Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev 91(3):1071–1121. doi:10.1152/physrev.00038.2010

    CAS  PubMed  Google Scholar 

  65. Kerbel RS, Viloria-Petit A, Klement G, Rak J (2000) ‘Accidental’ anti-angiogenic drugs. anti-oncogene directed signal transduction inhibitors and conventional chemotherapeutic agents as examples. Eur J Cancer 36(10):1248–1257

    CAS  PubMed  Google Scholar 

  66. Rak J, Yu JL (2004) Oncogenes and tumor angiogenesis: the question of vascular “supply” and vascular “demand”. Semin Cancer Biol 14(2):93–104

    CAS  PubMed  Google Scholar 

  67. Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, Fanning S, Zimmermann J, Lydon NB (1996) Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med 2(5):561–566

    CAS  PubMed  Google Scholar 

  68. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L (2001) Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 344(11):783–792. doi:10.1056/NEJM200103153441101

    CAS  PubMed  Google Scholar 

  69. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, Naoki K, Sasaki H, Fujii Y, Eck MJ, Sellers WR, Johnson BE, Meyerson M (2004) EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304(5676):1497–1500. doi:10.1126/science.1099314

    CAS  PubMed  Google Scholar 

  70. Ciardiello F, Tortora G (2008) EGFR antagonists in cancer treatment. N Engl J Med 358(11):1160–1174. doi:10.1056/NEJMra0707704

    CAS  PubMed  Google Scholar 

  71. Bottos A, Martini M, Di Nicolantonio F, Comunanza V, Maione F, Minassi A, Appendino G, Bussolino F, Bardelli A (2012) Targeting oncogenic serine/threonine-protein kinase BRAF in cancer cells inhibits angiogenesis and abrogates hypoxia. Proc Natl Acad Sci USA 109(6):E353–E359. doi:10.1073/pnas.1105026109

    CAS  PubMed  Google Scholar 

  72. Ancrile BB, O’Hayer KM, Counter CM (2008) Oncogenic ras-induced expression of cytokines: a new target of anti-cancer therapeutics. Mol Interv 8(1):22–27. doi:10.1124/mi.8.1.6

    CAS  PubMed  Google Scholar 

  73. Richard DE, Berra E, Gothie E, Roux D, Pouyssegur J (1999) p42/p44 mitogen-activated protein kinases phosphorylate hypoxia-inducible factor 1alpha (HIF-1alpha) and enhance the transcriptional activity of HIF-1. J Biol Chem 274(46):32631–32637

    CAS  PubMed  Google Scholar 

  74. Berra E, Pages G, Pouyssegur J (2000) MAP kinases and hypoxia in the control of VEGF expression. Cancer Metastasis Rev 19(1–2):139–145

    CAS  PubMed  Google Scholar 

  75. Kevil CG, De Benedetti A, Payne DK, Coe LL, Laroux FS, Alexander JS (1996) Translational regulation of vascular permeability factor by eukaryotic initiation factor 4E: implications for tumor angiogenesis. Int J Cancer 65(6):785–790. doi:10.1002/(SICI)1097-0215(19960315)65:6<785:AID-IJC14>3.0.CO;2-3

    CAS  PubMed  Google Scholar 

  76. Nucera C, Porrello A, Antonello ZA, Mekel M, Nehs MA, Giordano TJ, Gerald D, Benjamin LE, Priolo C, Puxeddu E, Finn S, Jarzab B, Hodin RA, Pontecorvi A, Nose V, Lawler J, Parangi S (2010) B-Raf(V600E) and thrombospondin-1 promote thyroid cancer progression. Proc Natl Acad Sci USA 107(23):10649–10654

    CAS  PubMed  Google Scholar 

  77. Zerilli M, Zito G, Martorana A, Pitrone M, Cabibi D, Cappello F, Giordano C, Rodolico V (2010) BRAF(V600E) mutation influences hypoxia-inducible factor-1alpha expression levels in papillary thyroid cancer. Mod Pathol 23(8):1052–1060

    CAS  PubMed  Google Scholar 

  78. Sharma A, Trivedi NR, Zimmerman MA, Tuveson DA, Smith CD, Robertson GP (2005) Mutant V599EB-Raf regulates growth and vascular development of malignant melanoma tumors. Cancer Res 65(6):2412–2421

    CAS  PubMed  Google Scholar 

  79. Kumar SM, Yu H, Edwards R, Chen L, Kazianis S, Brafford P, Acs G, Herlyn M, Xu X (2007) Mutant V600E BRAF increases hypoxia inducible factor-1alpha expression in melanoma. Cancer Res 67(7):3177–3184

    CAS  PubMed  Google Scholar 

  80. Niault TS, Baccarini M (2010) Targets of Raf in tumorigenesis. Carcinogenesis 31(7):1165–1174

    CAS  PubMed  Google Scholar 

  81. Kranenburg O, Gebbink MF, Voest EE (2004) Stimulation of angiogenesis by Ras proteins. Biochim Biophys Acta 1654(1):23–37. doi:10.1016/j.bbcan.2003.09.004

    CAS  PubMed  Google Scholar 

  82. Rak J, Mitsuhashi Y, Bayko L, Filmus J, Shirasawa S, Sasazuki T, Kerbel RS (1995) Mutant ras oncogenes upregulate VEGF/VPF expression: implications for induction and inhibition of tumor angiogenesis. Cancer Res 55(20):4575–4580

    CAS  PubMed  Google Scholar 

  83. Grugel S, Finkenzeller G, Weindel K, Barleon B, Marme D (1995) Both v-Ha-Ras and v-Raf stimulate expression of the vascular endothelial growth factor in NIH 3T3 cells. J Biol Chem 270(43):25915–25919

    CAS  PubMed  Google Scholar 

  84. Matsuo Y, Campbell PM, Brekken RA, Sung B, Ouellette MM, Fleming JB, Aggarwal BB, Der CJ, Guha S (2009) K-Ras promotes angiogenesis mediated by immortalized human pancreatic epithelial cells through mitogen-activated protein kinase signaling pathways. Mol Cancer Res 7(6):799–808. doi:10.1158/1541-7786.MCR-08-0577

    CAS  PubMed  Google Scholar 

  85. Mizukami Y, Jo WS, Duerr EM, Gala M, Li J, Zhang X, Zimmer MA, Iliopoulos O, Zukerberg LR, Kohgo Y, Lynch MP, Rueda BR, Chung DC (2005) Induction of interleukin-8 preserves the angiogenic response in HIF-1alpha-deficient colon cancer cells. Nat Med 11(9):992–997. doi:10.1038/nm1294

    CAS  PubMed  Google Scholar 

  86. Zabrenetzky V, Harris CC, Steeg PS, Roberts DD (1994) Expression of the extracellular matrix molecule thrombospondin inversely correlates with malignant progression in melanoma, lung and breast carcinoma cell lines. Int J Cancer 59(2):191–195

    CAS  PubMed  Google Scholar 

  87. Gum R, Lengyel E, Juarez J, Chen JH, Sato H, Seiki M, Boyd D (1996) Stimulation of 92-kDa gelatinase B promoter activity by ras is mitogen-activated protein kinase kinase 1-independent and requires multiple transcription factor binding sites including closely spaced PEA3/ets and AP-1 sequences. J Biol Chem 271(18):10672–10680

    CAS  PubMed  Google Scholar 

  88. Sato H, Kida Y, Mai M, Endo Y, Sasaki T, Tanaka J, Seiki M (1992) Expression of genes encoding type IV collagen-degrading metalloproteinases and tissue inhibitors of metalloproteinases in various human tumor cells. Oncogene 7(1):77–83

    CAS  PubMed  Google Scholar 

  89. Konishi T, Huang CL, Adachi M, Taki T, Inufusa H, Kodama K, Kohno N, Miyake M (2000) The K-ras gene regulates vascular endothelial growth factor gene expression in non-small cell lung cancers. Int J Oncol 16(3):501–511

    CAS  PubMed  Google Scholar 

  90. Ikeda N, Nakajima Y, Sho M, Adachi M, Huang CL, Iki K, Kanehiro H, Hisanaga M, Nakano H, Miyake M (2001) The association of K-ras gene mutation and vascular endothelial growth factor gene expression in pancreatic carcinoma. Cancer 92(3):488–499. doi:10.1002/1097-0142(20010801)92:3<488:AID-CNCR1347>3.0.CO;2-F

    CAS  PubMed  Google Scholar 

  91. Okada F, Rak JW, Croix BS, Lieubeau B, Kaya M, Roncari L, Shirasawa S, Sasazuki T, Kerbel RS (1998) Impact of oncogenes in tumor angiogenesis: mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells. Proc Natl Acad Sci USA 95(7):3609–3614

    CAS  PubMed  Google Scholar 

  92. Skinner HD, Zheng JZ, Fang J, Agani F, Jiang BH (2004) Vascular endothelial growth factor transcriptional activation is mediated by hypoxia-inducible factor 1alpha, HDM2, and p70S6K1 in response to phosphatidylinositol 3-kinase/AKT signaling. J Biol Chem 279(44):45643–45651. doi:10.1074/jbc.M404097200

    CAS  PubMed  Google Scholar 

  93. Hudson CC, Liu M, Chiang GG, Otterness DM, Loomis DC, Kaper F, Giaccia AJ, Abraham RT (2002) Regulation of hypoxia-inducible factor 1alpha expression and function by the mammalian target of rapamycin. Mol Cell Biol 22(20):7004–7014

    CAS  PubMed  Google Scholar 

  94. Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin WG Jr (2003) TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell 4(2):147–158. doi:S1535610803001879

    CAS  PubMed  Google Scholar 

  95. Qayum N, Muschel RJ, Im JH, Balathasan L, Koch CJ, Patel S, McKenna WG, Bernhard EJ (2009) Tumor vascular changes mediated by inhibition of oncogenic signaling. Cancer Res 69(15):6347–6354. doi:10.1158/0008-5472.CAN-09-0657

    CAS  PubMed  Google Scholar 

  96. Schnell CR, Stauffer F, Allegrini PR, O’Reilly T, McSheehy PM, Dartois C, Stumm M, Cozens R, Littlewood-Evans A, Garcia-Echeverria C, Maira SM (2008) Effects of the dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 on the tumor vasculature: implications for clinical imaging. Cancer Res 68(16):6598–6607. doi:10.1158/0008-5472.CAN-08-1044

    CAS  PubMed  Google Scholar 

  97. Baudino TA, McKay C, Pendeville-Samain H, Nilsson JA, Maclean KH, White EL, Davis AC, Ihle JN, Cleveland JL (2002) c-Myc is essential for vasculogenesis and angiogenesis during development and tumor progression. Genes Dev 16(19):2530–2543. doi:10.1101/gad.1024602

    CAS  PubMed  Google Scholar 

  98. Janz A, Sevignani C, Kenyon K, Ngo CV, Thomas-Tikhonenko A (2000) Activation of the myc oncoprotein leads to increased turnover of thrombospondin-1 mRNA. Nucleic Acids Res 28(11):2268–2275

    CAS  PubMed  Google Scholar 

  99. Pal S, Datta K, Mukhopadhyay D (2001) Central role of p53 on regulation of vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) expression in mammary carcinoma. Cancer Res 61(18):6952–6957

    CAS  PubMed  Google Scholar 

  100. Ueba T, Nosaka T, Takahashi JA, Shibata F, Florkiewicz RZ, Vogelstein B, Oda Y, Kikuchi H, Hatanaka M (1994) Transcriptional regulation of basic fibroblast growth factor gene by p53 in human glioblastoma and hepatocellular carcinoma cells. Proc Natl Acad Sci USA 91(19):9009–9013

    CAS  PubMed  Google Scholar 

  101. Dameron KM, Volpert OV, Tainsky MA, Bouck N (1994) Control of angiogenesis in fibroblasts by p53 regulation of thrombospondin-1. Science 265(5178):1582–1584

    CAS  PubMed  Google Scholar 

  102. Giuriato S, Ryeom S, Fan AC, Bachireddy P, Lynch RC, Rioth MJ, van Riggelen J, Kopelman AM, Passegue E, Tang F, Folkman J, Felsher DW (2006) Sustained regression of tumors upon MYC inactivation requires p53 or thrombospondin-1 to reverse the angiogenic switch. Proc Natl Acad Sci USA 103(44):16266–16271. doi:10.1073/pnas.0608017103

    CAS  PubMed  Google Scholar 

  103. Su F, Pascal LE, Xiao W, Wang Z (2010) Tumor suppressor U19/EAF2 regulates thrombospondin-1 expression via p53. Oncogene 29(3):421–431. doi:10.1038/onc.2009.326

    CAS  PubMed  Google Scholar 

  104. Pore N, Jiang Z, Gupta A, Cerniglia G, Kao GD, Maity A (2006) EGFR tyrosine kinase inhibitors decrease VEGF expression by both hypoxia-inducible factor (HIF)-1-independent and HIF-1-dependent mechanisms. Cancer Res 66(6):3197–3204. doi:10.1158/0008-5472.CAN-05-3090

    CAS  PubMed  Google Scholar 

  105. Ciardiello F, Caputo R, Bianco R, Damiano V, Fontanini G, Cuccato S, De Placido S, Bianco AR, Tortora G (2001) Inhibition of growth factor production and angiogenesis in human cancer cells by ZD1839 (Iressa), a selective epidermal growth factor receptor tyrosine kinase inhibitor. Clin Cancer Res 7(5):1459–1465

    CAS  PubMed  Google Scholar 

  106. Cerniglia GJ, Pore N, Tsai JH, Schultz S, Mick R, Choe R, Xing X, Durduran T, Yodh AG, Evans SM, Koch CJ, Hahn SM, Quon H, Sehgal CM, Lee WM, Maity A (2009) Epidermal growth factor receptor inhibition modulates the microenvironment by vascular normalization to improve chemotherapy and radiotherapy efficacy. PLoS One 4(8):e6539. doi:10.1371/journal.pone.0006539

    PubMed  Google Scholar 

  107. Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK (2002) Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature 416(6878):279–280. doi:10.1038/416279b

    CAS  PubMed  Google Scholar 

  108. Ellis LM, Hicklin DJ (2009) Resistance to targeted therapies: refining anticancer Therapy in the Era of molecular oncology. Clin Cancer Res 15(24):7471–7478. doi:10.1158/1078-0432.CCR-09-1070

    CAS  PubMed  Google Scholar 

  109. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674. doi:10.1016/j.cell.2011.02.013

    CAS  PubMed  Google Scholar 

  110. Allegra CJ, Yothers G, O’Connell MJ, Sharif S, Petrelli NJ, Colangelo LH, Atkins JN, Seay TE, Fehrenbacher L, Goldberg RM, O’Reilly S, Chu L, Azar CA, Lopa S, Wolmark N (2011) Phase III trial assessing bevacizumab in stages II and III carcinoma of the colon: results of NSABP protocol C-08. J Clin Oncol 29(1):11–16. doi:10.1200/JCO.2010.30.0855

    CAS  PubMed  Google Scholar 

  111. Kerr DJ, Young AM (2011) Targeted therapies: bevacizumab–has it reached its final resting place? Nat Rev Clin Oncol 8(4):195–196. doi:10.1038/nrclinonc.2011.32

    CAS  PubMed  Google Scholar 

  112. Murukesh N, Dive C, Jayson GC (2010) Biomarkers of angiogenesis and their role in the development of VEGF inhibitors. Br J Cancer 102(1):8–18. doi:10.1038/sj.bjc.6605483

    CAS  PubMed  Google Scholar 

  113. Sessa C, Guibal A, Del Conte G, Ruegg C (2008) Biomarkers of angiogenesis for the development of antiangiogenic therapies in oncology: tools or decorations? Nat Clin Pract Oncol 5(7):378–391. doi:10.1038/ncponc1150

    CAS  PubMed  Google Scholar 

  114. Shojaei F (2012) Anti-angiogenesis therapy in cancer: current challenges and future perspectives. Cancer Lett 320(2):130–137. doi:10.1016/j.canlet.2012.03.008

    CAS  PubMed  Google Scholar 

  115. Jubb AM, Hurwitz HI, Bai W, Holmgren EB, Tobin P, Guerrero AS, Kabbinavar F, Holden SN, Novotny WF, Frantz GD, Hillan KJ, Koeppen H (2006) Impact of vascular endothelial growth factor-A expression, thrombospondin-2 expression, and microvessel density on the treatment effect of bevacizumab in metastatic colorectal cancer. J Clin Oncol 24(2):217–227. doi:10.1200/JCO.2005.01.5388

    CAS  PubMed  Google Scholar 

  116. Ince WL, Jubb AM, Holden SN, Holmgren EB, Tobin P, Sridhar M, Hurwitz HI, Kabbinavar F, Novotny WF, Hillan KJ, Koeppen H (2005) Association of k-ras, b-raf, and p53 status with the treatment effect of bevacizumab. J Natl Cancer Inst 97(13):981–989. doi:10.1093/jnci/dji174

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

A warm thanks to Prof. Nancy E. Hynes, Dr. Jason W. Gill, and Dr. Alessio Noghero for critical reading and suggestions. The research leading to these results has received funding from the European Community’s Seventh Framework Programme under Grant agreement no. 259015 COLTHERES; AIRC 2010 Special Program Molecular Clinical Oncology 5xMille, Project no. 9970; Intramural Grant—5xmille 2008 Fondazione Piemontese per la Ricerca sul Cancro—ONLUS, AIRC IG Grant no. 12812. A. Bottos was supported by the Swiss Science Foundation (#310030_138417).

Conflict of interest

A. Bardelli owns stock in, and has received consultancy fees, from Horizon Discovery.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alberto Bardelli.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bottos, A., Bardelli, A. Oncogenes and angiogenesis: a way to personalize anti-angiogenic therapy?. Cell. Mol. Life Sci. 70, 4131–4140 (2013). https://doi.org/10.1007/s00018-013-1331-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-013-1331-3

Keywords

Navigation