1932

Abstract

RAS proteins play a major, causal role in many human cancers. No therapies have been developed for these cancers because the RAS protein has been considered undruggable given that it has no accessible pocket to which a drug could bind with high affinity, and the mutant proteins that cause cancer are virtually identical to their essential, wild-type counterparts. New technologies in drug development, such as nuclear magnetic resonance–based fragment screening and covalent tethering, and new insights into RAS structure and function have changed this perception and facilitated the development of several drug candidates.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-cancerbio-050216-122010
2018-03-04
2024-04-23
Loading full text...

Full text loading...

/deliver/fulltext/cancerbio/2/1/annurev-cancerbio-050216-122010.html?itemId=/content/journals/10.1146/annurev-cancerbio-050216-122010&mimeType=html&fmt=ahah

Literature Cited

  1. Ahmadian MR, Zor T, Vogt D, Kabsch W, Selinger Z. et al. 1999. Guanosine triphosphatase stimulation of oncogenic Ras mutants. PNAS 96:7065–70 [Google Scholar]
  2. Bos JL, Rehmann H, Wittinghofer A. 2007. GEFs and GAPs: critical elements in the control of small G proteins. Cell 129:865–77 [Google Scholar]
  3. Bourne HR, Sanders DA, McCormick F. 1991. The GTPase superfamily: conserved structure and molecular mechanism. Nature 349:117–27 [Google Scholar]
  4. Cetin M, Evenson WE, Gross GG, Jalali-Yazdi F, Krieger D. et al. 2017. RasIns: genetically encoded intrabodies of activated Ras proteins. J. Mol. Biol. 429:562–73 [Google Scholar]
  5. Clark GJ, Drugan JK, Terrell RS, Bradham C, Der CJ. et al. 1996. Peptides containing a consensus Ras binding sequence from Raf-1 and the GTPase activating protein NF1 inhibit Ras function. PNAS 93:1577–81 [Google Scholar]
  6. Cox AD, Der CJ, Philips MR. 2015. Targeting RAS membrane association: back to the future for anti-RAS drug discovery?. Clin. Cancer Res. 21:1819–27 [Google Scholar]
  7. Feramisco JR, Clark R, Wong G, Arnheim N, Milley R, McCormick F. 1985. Transient reversion of ras oncogene-induced cell transformation by antibodies specific for amino acid 12 of ras protein. Nature 314:639–42 [Google Scholar]
  8. Hancock JF, Magee AI, Childs JE, Marshall CJ. 1989. All ras proteins are polyisoprenylated but only some are palmitoylated. Cell 57:1167–77 [Google Scholar]
  9. Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. 2015. Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol. Cancer Res. 13:1325–35 [Google Scholar]
  10. Lai AC, Toure M, Hellerschmied D, Salami J, Jaime-Figueroa S. et al. 2016. Modular PROTAC design for the degradation of oncogenic BCR-ABL. Angew. Chem. Int. Ed. 55:807–10 [Google Scholar]
  11. Leshchiner ES, Parkhitko A, Bird GH, Luccarelli J, Bellairs JA. et al. 2015. Direct inhibition of oncogenic KRAS by hydrocarbon-stapled SOS1 helices. PNAS 112:1761–66 [Google Scholar]
  12. Maurer T, Garrenton LS, Oh A, Pitts K, Anderson DJ. et al. 2012. Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity. PNAS 109:5299–304 [Google Scholar]
  13. Mott HR, Owen D. 2015. Structures of Ras superfamily effector complexes: What have we learnt in two decades?. Crit. Rev. Biochem. Mol. Biol. 50:85–133 [Google Scholar]
  14. Nan X, Tamguney TM, Collisson EA, Lin LJ, Pitt C. et al. 2015. Ras-GTP dimers activate the Mitogen-Activated Protein Kinase (MAPK) pathway. PNAS 112:7996–8001 [Google Scholar]
  15. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. 2013. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 503:548–51 [Google Scholar]
  16. Ostrem JM, Shokat KM. 2016. Direct small-molecule inhibitors of KRAS: from structural insights to mechanism-based design. Nat. Rev. Drug. Discov. 15:771–85 [Google Scholar]
  17. Patricelli MP, Janes MR, Li LS, Hansen R, Peters U. et al. 2016. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov 6:316–29 [Google Scholar]
  18. Prior IA, Lewis PD, Mattos C. 2012. A comprehensive survey of Ras mutations in cancer. Cancer Res 72:2457–67 [Google Scholar]
  19. Quinlan MP, Quatela SE, Philips MR, Settleman J. 2008. Activated Kras, but not Hras or Nras, may initiate tumors of endodermal origin via stem cell expansion. Mol. Cell Biol. 28:2659–74 [Google Scholar]
  20. Scheffzek K, Ahmadian MR, Kabsch W, Wiesmuller L, Lautwein A. et al. 1997. The Ras-RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants. Science 277:333–38 [Google Scholar]
  21. Shuker SB, Hajduk PJ, Meadows RP, Fesik SW. 1996. Discovering high-affinity ligands for proteins: SAR by NMR. Science 274:1531–34 [Google Scholar]
  22. Spencer-Smith R, Koide A, Zhou Y, Eguchi RR, Sha F. et al. 2017. Inhibition of RAS function through targeting an allosteric regulatory site. Nat. Chem. Biol. 13:62–68 [Google Scholar]
  23. Stephen AG, Esposito D, Bagni RK, McCormick F. 2014. Dragging Ras back in the ring. Cancer Cell 25:272–81 [Google Scholar]
  24. Sun Q, Burke JP, Phan J, Burns MC, Olejniczak ET. et al. 2012. Discovery of small molecules that bind to K-Ras and inhibit Sos-mediated activation. Angew. Chem. Int. Ed. 51:6140–43 [Google Scholar]
  25. Sung PJ, Tsai FD, Vais H, Court H, Yang J. et al. 2013. Phosphorylated K-Ras limits cell survival by blocking Bcl-xL sensitization of inositol trisphosphate receptors. PNAS 110:20593–98 [Google Scholar]
  26. Tanaka T, Rabbitts TH. 2010. Interfering with RAS–effector protein interactions prevent RAS-dependent tumour initiation and causes stop–start control of cancer growth. Oncogene 29:6064–70 [Google Scholar]
  27. Upadhyaya P, Qian Z, Selner NG, Clippinger SR, Wu Z. et al. 2015. Inhibition of Ras signaling by blocking Ras–effector interactions with cyclic peptides. Angew. Chem. Int. Ed. 54:7602–6 [Google Scholar]
  28. Villalonga P, Lopez-Alcala C, Bosch M, Chiloeches A, Rocamora N. et al. 2001. Calmodulin binds to K-Ras, but not to H- or N-Ras, and modulates its downstream signaling. Mol. Cell Biol. 21:7345–54 [Google Scholar]
  29. Wang MT, Holderfield M, Galeas J, Delrosario R, To MD. et al. 2015. K-Ras promotes tumorigenicity through suppression of non-canonical Wnt signaling. Cell 163:1237–51 [Google Scholar]
  30. Wang T, Yu H, Hughes NW, Liu B, Kendirli A. et al. 2017. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168:890–903.e815 [Google Scholar]
  31. Welsch ME, Kaplan A, Chambers JM, Stokes ME, Bos PH. et al. 2017. Multivalent small-molecule pan-RAS inhibitors. Cell 168:878–89.e829 [Google Scholar]
  32. Willumsen BM, Christensen A, Hubbert NL, Papageorge AG, Lowy DR. 1984. The p21 ras C-terminus is required for transformation and membrane association. Nature 310:583–86 [Google Scholar]
  33. Zhou Y, Prakash P, Liang H, Cho KJ, Gorfe AA, Hancock JF. 2017. Lipid-sorting specificity encoded in K-Ras membrane anchor regulates signal output. Cell 168:239–51.e216 [Google Scholar]
  34. Zimmermann G, Papke B, Ismail S, Vartak N, Chandra A. et al. 2013. Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature 497:638–42 [Google Scholar]
/content/journals/10.1146/annurev-cancerbio-050216-122010
Loading
/content/journals/10.1146/annurev-cancerbio-050216-122010
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error