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:

Small-molecule modulation of Ras signaling

Abstract

Despite intense efforts in pharmaceutical industry and academia, a therapeutic grip on oncogenic Ras proteins has remained elusive. Mutated Ras is associated with 20−30% of all human cancers often not responsive to established therapies. In particular, K-Ras, the most frequently mutated Ras isoform, is considered one of the most important but 'undruggable' targets in cancer research. Recently, new cavities on Ras for small-molecule ligands were identified, and selective direct targeting of mutated K-RasG12C has become possible for what is to our knowledge the first time. In addition, impairment of Ras spatial organization, in particular via targeting the prenyl-binding Ras chaperone PDEδ, has opened a fresh perspective in anticancer research. These recent advances fuel hopes for the development of new drugs targeting Ras.

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: Regulation of Ras activity.
Figure 2: Strategies for targeting Ras.
Figure 3: Structural basis for orthosteric inhibition of Ras-GEF interactions.
Figure 4: Covalent inhibitors targeting Cys12 of mutated K-RasG12C.
Figure 5: Structural basis for orthosteric inhibition of Ras−effector interactions.
Figure 6: Post-translational processing of Ras.
Figure 7: Structural basis for inhibition of the K-Ras−PDEδ interaction.
Figure 8: Inhibition of Rab-effector interactions.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Forbes, S.A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Prior, I.A., Lewis, P.D. & Mattos, C.A. Comprehensive survey of Ras mutations in cancer. Cancer Res. 72, 2457–2467 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Helwick, C. Targeting KRAS in GI Cancers: the hunt for the Holy Grail in cancer research. The ASCO Post, http://www.ascopost.com/issues/april-15-2012/targeting-kras-in-gi-cancers-the-hunt-for-the-holy-grail-in-cancer-research.aspx (2012).

  4. Wennerberg, K., Rossman, K.L. & Der, C.J. The Ras superfamily at a glance. J. Cell Sci. 118, 843–846 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Cherfils, J. & Zeghouf, M. Regulation of Small GTPases by GEFs, GAPs, and GDIs. Physiol. Rev. 93, 269–309 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Vetter, I.R. & Wittinghofer, A. The guanine nucleotide-binding switch in three dimensions. Science 294, 1299–1304 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Prior, I.A. & Hancock, J.F. Ras trafficking, localization and compartmentalized signalling. Semin. Cell Dev. Biol. 23, 145–153 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Pylayeva-Gupta, Y., Grabocka, E. & Bar-Sagi, D. RAS oncogenes: weaving a tumorigenic web. Nat. Rev. Cancer 11, 761–774 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Baines, A.T., Xu, D. & Der, C.J. Inhibition of Ras for cancer treatment: the search continues. Future Med. Chem. 3, 1787–1808 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Lito, P., Rosen, N. & Solit, D.B. Tumor adaptation and resistance to RAF inhibitors. Nat. Med. 19, 1401–1409 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Berndt, N., Hamilton, A.D. & Sebti, S.M. Targeting protein prenylation for cancer therapy. Nat. Rev. Cancer 11, 775–791 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang, Y., Kaiser, C.E., Frett, B. & Li, H.-y. Targeting mutant KRAS for anticancer therapeutics: a review of novel small molecule modulators. J. Med. Chem. 56, 5219–5230 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wang, W., Fang, G. & Rudolph, J. Ras inhibition via direct Ras binding—is there a path forward? Bioorg. Med. Chem. Lett. 22, 5766–5776 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Noonan, T., Brown, N., Dudycz, L. & Wright, G. Interaction of GTP derivatives with cellular and oncogenic ras-p21 proteins. J. Med. Chem. 34, 1302–1307 (1991).

    Article  CAS  PubMed  Google Scholar 

  15. Wolin, R., et al. Synthesis and evaluation of pyrazolo[3,4-b]quinoline ribofuranosides and their derivatives as inhibitors of oncogenic Ras. Bioorg. Med. Chem. Lett. 6, 195–200 (1996).

    Article  CAS  Google Scholar 

  16. Becher, I. et al. Affinity profiling of the cellular kinome for the nucleotide cofactors ATP, ADP, and GTP. ACS Chem. Biol. 8, 599–607 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. John, J. et al. Kinetic and structural analysis of the Mg2+-binding site of the guanine nucleotide-binding protein p21H-ras. J. Biol. Chem. 268, 923–929 (1993).

    CAS  PubMed  Google Scholar 

  18. Freedman, T.S. et al. A Ras-induced conformational switch in the Ras activator Son of sevenless. Proc. Natl. Acad. Sci. USA 103, 16692–16697 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Taveras, A.G. et al. Ras oncoprotein inhibitors: the discovery of potent, ras nucleotide exchange inhibitors and the structural determination of a drug–protein complex. Bioorg. Med. Chem. 5, 125–133 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Ganguly, A.K. et al. Interaction of a novel GDP exchange inhibitor with the Ras protein. Biochemistry 37, 15631–15637 (1998).

    Article  CAS  PubMed  Google Scholar 

  21. Peri, F. et al. Design, synthesis and biological evaluation of sugar-derived Ras inhibitors. ChemBioChem 6, 1839–1848 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Sacco, E. et al. Binding properties and biological characterization of new sugar-derived Ras ligands. Med. Chem. Commun. 2, 396 (2011).

    Article  CAS  Google Scholar 

  23. Patgiri, A., Yadav, K.K., Arora, P.S. & Bar-Sagi, D. An orthosteric inhibitor of the Ras-Sos interaction. Nat. Chem. Biol. 7, 585–587 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sacco, E. et al. Novel RasGRF1-derived Tat-fused peptides inhibiting Ras-dependent proliferation and migration in mouse and human cancer cells. Biotechnol. Adv. 30, 233–243 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Gorfe, A.A., Grant, B.J. & McCammon, J.A. Mapping the nucleotide and isoform-dependent structural and dynamical features of Ras proteins. Structure 16, 885–896 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Grant, B.J. et al. Novel allosteric sites on Ras for lead generation. PLoS ONE 6, e25711 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hocker, H.J. et al. Andrographolide derivatives inhibit guanine nucleotide exchange and abrogate oncogenic Ras function. Proc. Natl. Acad. Sci. USA 110, 10201–10206 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hajduk, P.J. & Greer, J. A decade of fragment-based drug design: strategic advances and lessons learned. Nat. Rev. Drug Discov. 6, 211–219 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Maurer, T. et al. Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity. Proc. Natl. Acad. Sci. USA 109, 5299–5304 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sun, Q. et al. Discovery of small molecules that bind to K-Ras and inhibit Sos-mediated activation. Angew. Chem. Int. Ed. Engl. 51, 6140–6143 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schöpel, M. et al. Bisphenol A binds to Ras proteins and competes with guanine nucleotide exchange: implications for GTPase-selective antagonists. J. Med. Chem. 56, 9664–9672 (2013).

    Article  PubMed  CAS  Google Scholar 

  32. Burns, M.C. et al. Approach for targeting Ras with small molecules that activate SOS-mediated nucleotide exchange. Proc. Natl. Acad. Sci. USA 111, 3401–3406 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lim, S.M. et al. Therapeutic Targeting of Oncogenic K-Ras by a Covalent Catalytic Site Inhibitor. Angew. Chem. Int. Ed. Engl. 53, 199–204 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Ostrem, J.M., Peters, U., Sos, M.L., Wells, J.A. & Shokat, K.M. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 503, 548–551 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Geyer, M. et al. Conformational Transitions in p21 Ras and in its complexes with the effector protein Raf-RBD and the GTPase activating protein GAP. Biochemistry 35, 10308–10320 (1996).

    Article  CAS  PubMed  Google Scholar 

  36. Spoerner, M., Herrmann, C., Vetter, I.R., Kalbitzer, H.R. & Wittinghofer, A. Dynamic properties of the Ras switch I region and its importance for binding to effectors. Proc. Natl. Acad. Sci. USA 98, 4944–4949 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Spoerner, M., Wittinghofer, A. & Kalbitzer, H.R. Perturbation of the conformational equilibria in Ras by selective mutations as studied by 31P NMR spectroscopy. FEBS Lett. 578, 305–310 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Liao, J. et al. Two conformational states of Ras GTPase exhibit differential GTP-binding kinetics. Biochem. Biophys. Res. Commun. 369, 327–332 (2008).

    Article  CAS  PubMed  Google Scholar 

  39. Spoerner, M., Graf, T., Konig, B. & Kalbitzer, H.R. A novel mechanism for the modulation of the Ras-effector interaction by small molecules. Biochem. Biophys. Res. Commun. 334, 709–713 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Rosnizeck, I.C. et al. Stabilizing a weak binding state for effectors in the human Ras protein by cyclen complexes. Angew. Chem. Int. Ed. Engl. 49, 3830–3833 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Rosnizeck, I.C. et al. Metal-bis(2-picolyl)amine complexes as state 1(T) inhibitors of activated Ras protein. Angew. Chem. Int. Ed. Engl. 51, 10647–10651 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Tanaka, T., Williams, R.L. & Rabbitts, T.H. Tumour prevention by a single antibody domain targeting the interaction of signal transduction proteins with RAS. EMBO J. 26, 3250–3259 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tanaka, T. & Rabbitts, T.H. Interfering with RAS-effector protein interactions prevent RAS-dependent tumour initiation and causes stop-start control of cancer growth. Oncogene 29, 6064–6070 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Barnard, D., Sun, H., Baker, L. & Marshall, M.S. In vitro inhibition of Ras–Raf association by short peptides. Biochem. Biophys. Res. Commun. 247, 176–180 (1998).

    Article  CAS  PubMed  Google Scholar 

  45. Wu, X., Upadhyaya, P., Villalona-Calero, M.A., Briesewitz, R. & Pei, D. Inhibition of Ras-effector interactions by cyclic peptides. Med. Chem. Commun. 4, 378 (2013).

    Article  Google Scholar 

  46. Pasricha, P.J. et al. The effects of sulindac on colorectal proliferation and apoptosis in familial adenomatous polyposis. Gastroenterology 109, 994–998 (1995).

    Article  CAS  PubMed  Google Scholar 

  47. Herrmann, C. et al. Sulindac sulfide inhibits Ras signaling. Oncogene 17, 1769–1776 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Müller, O. et al. Identification of potent Ras signaling inhibitors by pathway-selective phenotype-based screening. Angew. Chem. Int. Ed. Engl. 43, 450–454 (2004).

    Article  PubMed  CAS  Google Scholar 

  49. Waldmann, H. et al. Sulindac-derived Ras pathway inhibitors target the Ras-Raf interaction and downstream effectors in the Ras pathway. Angew. Chem. Int. Ed. Engl. 43, 454–458 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. González-Pérez, V. et al. Genetic and functional characterization of putative Ras/Raf interaction inhibitors in C. elegans and mammalian cells. J. Mol. Signal. 5, 2 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Kato-Stankiewicz, J. et al. Inhibitors of Ras/Raf-1 interaction identified by two-hybrid screening revert Ras-dependent transformation phenotypes in human cancer cells. Proc. Natl. Acad. Sci. USA 99, 14398–14403 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lu, Y. et al. Solution phase parallel synthesis and evaluation of MAPK inhibitory activities of close structural analogues of a Ras pathway modulator. Bioorg. Med. Chem. Lett. 14, 3957–3962 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Skobeleva, N., Menon, S., Weber, L., Golemis, E.A. & Khazak, V. In vitro and in vivo synergy of MCP compounds with mitogen-activated protein kinase pathway- and microtubule-targeting inhibitors. Mol. Cancer Ther. 6, 898–906 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Shima, F. et al. Structural basis for conformational dynamics of GTP-bound Ras protein. J. Biol. Chem. 285, 22696–22705 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Shima, F. et al. In silico discovery of small-molecule Ras inhibitors that display antitumor activity by blocking the Ras-effector interaction. Proc. Natl. Acad. Sci. USA 110, 8182–8187 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ahearn, I.M., Haigis, K., Bar-Sagi, D. & Philips, M.R. Regulating the regulator: post-translational modification of RAS. Nat. Rev. Mol. Cell Biol. 13, 39–51 (2012).

    Article  CAS  Google Scholar 

  57. Whyte, D.B. et al. K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J. Biol. Chem. 272, 14459–14464 (1997).

    Article  CAS  PubMed  Google Scholar 

  58. Lobell, R.B. et al. Preclinical and clinical pharmacodynamic assessment of L-778,123, a dual inhibitor of farnesyl:protein transferase and geranylgeranyl:protein transferase type-I. Mol. Cancer Ther. 1, 747–758 (2002).

    CAS  PubMed  Google Scholar 

  59. Gnant, M. et al. Endocrine therapy plus zoledronic acid in premenopausal breast cancer. N. Engl. J. Med. 360, 679–691 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Jahnke, W. et al. Allosteric non-bisphosphonate FPPS inhibitors identified by fragment-based discovery. Nat. Chem. Biol. 6, 660–666 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Winter-Vann, A.M. & Casey, P.J. Opinion: post-prenylation-processing enzymes as new targets in oncogenesis. Nat. Rev. Cancer 5, 405–412 (2005).

    Article  CAS  PubMed  Google Scholar 

  62. Go, M.-L. et al. Amino derivatives of indole as potent inhibitors of isoprenylcysteine carboxyl methyltransferase. J. Med. Chem. 53, 6838–6850 (2010).

    Article  CAS  PubMed  Google Scholar 

  63. Wahlstrom, A.M. et al. Inactivating Icmt ameliorates K-RAS–induced myeloproliferative disease. Blood 112, 1357–1365 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Riely, G.J. et al. A phase II trial of salirasib in patients with lung adenocarcinomas with KRAS mutations. J. Thorac. Oncol. 6, 1435–1437 (2011).

    Article  PubMed  Google Scholar 

  65. Laheru, D. et al. Integrated preclinical and clinical development of S-trans, trans-farnesylthiosalicylic acid (FTS, Salirasib) in pancreatic cancer. Invest. New Drugs 30, 2391–2399 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Campbell, P.M. et al. TLN-4601 suppresses growth and induces apoptosis of pancreatic carcinoma cells through inhibition of Ras-ERK MAPK signaling. J. Mol. Signal. 5, 18 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Rocks, O. et al. An acylation cycle regulates localization and activity of palmitoylated Ras isoforms. Science 307, 1746–1752 (2005).

    Article  CAS  PubMed  Google Scholar 

  68. Rocks, O., Peyker, A. & Bastiaens Philippe, I.H. Spatio-temporal segregation of Ras signals: one ship, three anchors, many harbors. Curr. Opin. Cell Biol. 18, 351–357 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Rocks, O. et al. The palmitoylation machinery is a spatially organizing system for peripheral membrane proteins. Cell 141, 458–471 (2010).

    Article  CAS  PubMed  Google Scholar 

  70. Dekker, F.J. et al. Small-molecule inhibition of APT1 affects Ras localization and signaling. Nat. Chem. Biol. 6, 449–456 (2010).

    Article  CAS  PubMed  Google Scholar 

  71. Rusch, M. et al. Identification of acyl protein thioesterases 1 and 2 as the cellular targets of the Ras-signaling modulators palmostatin B and M. Angew. Chem. Int. Edn Engl. 50, 9838–9842 (2011).

    Article  CAS  Google Scholar 

  72. Hedberg, C. et al. Development of highly potent inhibitors of the Ras-targeting human acyl protein thioesterases based on substrate similarity design. Angew. Chem. Int. Edn Engl. 50, 9832–9837 (2011).

    Article  CAS  Google Scholar 

  73. Adibekian, A. et al. Confirming target engagement for reversible inhibitors in vivo by kinetically tuned activity-based probes. J. Am. Chem. Soc. 134, 10345–10348 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chandra, A. et al. The GDI-like solubilizing factor PDEδ sustains the spatial organization and signalling of Ras family proteins. Nat. Cell Biol. 14, 148–158 (2012).

    Article  CAS  Google Scholar 

  75. Ismail, S.A. et al. Arl2-GTP and Arl3-GTP regulate a GDI-like transport system for farnesylated cargo. Nat. Chem. Biol. 7, 942–949 (2011).

    Article  CAS  PubMed  Google Scholar 

  76. Zimmermann, G. et al. Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature 497, 638–642 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Humbert, M.C. et al. ARL13B, PDE6D, and CEP164 form a functional network for INPP5E ciliary targeting. Proc. Natl. Acad. Sci. USA 109, 19691–19696 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Zhang, H., Constantine, R., Frederick, J.M. & Baehr, W. The prenyl-binding protein PrBP/δ: a chaperone participating in intracellular trafficking. Vision Res. 75, 19–25 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Thomas, S. et al. A homozygous PDE6D mutation in Joubert syndrome impairs targeting of farnesylated INPP5E protein to the primary cilium. Hum. Mutat. 35, 137–146 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Schmick, M. et al. KRas localizes to the plasma membrane by spatial cycles of solubilization, trapping and vesicular transport. Cell 157, 459–471 (2014).

    Article  CAS  PubMed  Google Scholar 

  81. Stephen, A.G., Esposito, D., Bagni, R.K. & McCormick, F. Dragging Ras back in the ring. Cancer Cell 25, 272–281 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Young, A., Lou, D. & McCormick, F. Oncogenic and wild-type Ras play divergent roles in the regulation of mitogen-activated protein kinase signaling. Cancer Discov. 3, 112–123 (2013).

    Article  CAS  PubMed  Google Scholar 

  83. Bentley, C. et al. A requirement for wild-type Ras isoforms in mutant KRas-driven signalling and transformation. Biochem. J. 452, 313–320 (2013).

    Article  CAS  PubMed  Google Scholar 

  84. To, M.D., Rosario, R.D., Westcott, P.M.K., Banta, K.L. & Balmain, A. Interactions between wild-type and mutant Ras genes in lung and skin carcinogenesis. Oncogene 32, 4028–4033 (2013).

    Article  CAS  PubMed  Google Scholar 

  85. Grabocka, E. et al. Wild-type H- and N-Ras promote mutant K-Ras–driven tumorigenesis by modulating the DNA damage response. Cancer Cell 25, 243–256 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Luo, F. et al. Wild-type K-ras has a tumour suppressor effect on carcinogen-induced murine colorectal adenoma formation. Int. J. Exp. Pathol. 95, 8–15 (2014).

    Article  CAS  PubMed  Google Scholar 

  87. Kleiner, R.E., Dumelin, C.E. & Liu, D.R. Small-molecule discovery from DNA-encoded chemical libraries. Chem. Soc. Rev. 40, 5707–5717 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Murakami, H., Ohta, A., Ashigai, H. & Suga, H. A highly flexible tRNA acylation method for non-natural polypeptide synthesis. Nat. Methods 3, 357–359 (2006).

    Article  CAS  PubMed  Google Scholar 

  89. Goto, Y. & Suga, H. Translation initiation with initiator tRNA charged with exotic peptides. J. Am. Chem. Soc. 131, 5040–5041 (2009).

    Article  CAS  PubMed  Google Scholar 

  90. Walensky, L.D. & Bird, G.H. Hydrocarbon-stapled peptides: principles, practice, and progress. J. Med. Chem. 10.1021/jm4011675 (2014).

  91. Kim, Y.-W., Grossmann, T.N. & Verdine, G.L. Synthesis of all-hydrocarbon stapled α-helical peptides by ring-closing olefin metathesis. Nat. Protoc. 6, 761–771 (2011).

    Article  CAS  PubMed  Google Scholar 

  92. Spiegel, J. et al. Direct targeting of Rab-GTPase-effector interactions. Angew. Chem. Int. Ed. Engl. 53, 2498–2503 (2014).

    Article  CAS  PubMed  Google Scholar 

  93. Ahmadian, M.R. et al. Guanosine triphosphatase stimulation of oncogenic Ras mutants. Proc. Natl. Acad. Sci. USA 96, 7065–7070 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Gail, R., Costisella, B., Ahmadian, M., Reza & Wittinghofer, A. Ras-mediated cleavage of a GTP Analogue by a novel mechanism. ChemBioChem 2, 570–575 (2001).

    Article  CAS  PubMed  Google Scholar 

  95. Zhang, H. et al. Deletion of PrBP δ impedes transport of GRK1 and PDE6 catalytic subunits to photoreceptor outer segments. Proc. Natl. Acad. Sci. USA 104, 8857–8862 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Prahallad, A. et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012).

    Article  CAS  PubMed  Google Scholar 

  97. Kamioka, Y., Yasuda, S., Fujita, Y., Aoki, K. & Matsuda, M. Multiple decisive phosphorylation sites for the negative feedback regulation of SOS1 via ERK. J. Biol. Chem. 285, 33540–33548 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Navas, C. et al. EGF receptor signaling is essential for K-Ras oncogene-driven pancreatic ductal adenocarcinoma. Cancer Cell 22, 318–330 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Nussinov, R., Tsai, C.-J. & Mattos, C. 'Pathway drug cocktail': targeting Ras signaling based on structural pathways. Trends Mol. Med. 19, 695–704 (2013).

    Article  PubMed  CAS  Google Scholar 

  100. Hayes, T.K. & Der, C.J. Mutant and wild-type Ras: co-conspirators in cancer. Cancer Discov. 3, 24–26 (2013).

    Article  CAS  PubMed  Google Scholar 

  101. Scheffzek, K. et al. The Ras–RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants. Science 277, 333–338 (1997).

    Article  CAS  PubMed  Google Scholar 

  102. Scheidig, A.J., Burmester, C. & Goody, R.S. The pre-hydrolysis state of p21ras in complex with GTP: new insights into the role of water molecules in the GTP hydrolysis reaction of ras-like proteins. Structure 7, 1311–1324 (1999).

    Article  CAS  PubMed  Google Scholar 

  103. Buhrman, G., Holzapfel, G., Fetics, S. & Mattos, C. Allosteric modulation of Ras positions Q61 for a direct role in catalysis. Proc. Natl. Acad. Sci. USA 107, 4931–4936 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Muraoka, S. et al. Crystal structures of the state 1 conformations of the GTP-bound H-Ras protein and its oncogenic G12V and Q61L mutants. FEBS Lett. 586, 1715–1718 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

J.S. acknowledges financial support by Fonds der Chemischen Industrie. P.M.C. is thankful to the Studienstiftung des deutschen Volkes for a fellowship. T.N.G. thanks the German Research Foundation (DFG, Emmy Noether program GR3592/2-1) for financial support.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Tom N Grossmann or Herbert Waldmann.

Ethics declarations

Competing interests

G.Z. and H.W. are inventors on a Max-Planck-Gesellschaft patent application concerning Ras-PDEδ inhibitors.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Spiegel, J., Cromm, P., Zimmermann, G. et al. Small-molecule modulation of Ras signaling. Nat Chem Biol 10, 613–622 (2014). https://doi.org/10.1038/nchembio.1560

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1560

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer