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:

Drugging the undruggable RAS: Mission Possible?

Key Points

  • Attempts to target RAS pharmaceutically have not yet yielded small molecules with sufficient potency and drug-like characteristics to be useful, but additional approaches are underway.

  • Inhibitors of RAS membrane interaction and subcellular localization remain attractive targets for therapeutic intervention. In this regard, recent discoveries of functionally relevant post-translational modifications of RAS, such as phosphorylation and ubiquitylation, suggest new opportunities to block RAS function.

  • Inhibitors of RAS effector signalling are currently centred on the RAF–MEK–ERK and PI3K–AKT–TOR pathways, with many inhibitors of components of these two pathways under clinical evaluation. The less well-studied effector pathways that lead to activation of RAL and RAC small GTPases are also promising targets.

  • Despite some setbacks owing to issues of reproducibility, functional screens for synthetic lethal interactors with oncogenic RAS remain an attractive approach to identify novel drug targets for RAS-driven cancers.

  • RAS-driven cancers become efficient scavengers of nutrients and may rely on pathways such as autophagy and macropinocytosis. Furthermore, oncogenic KRAS has a crucial role in altering tumour metabolism, including rewiring of both glucose- and glutamine-dependent metabolic pathways. These alterations could provide the opportunity for novel therapeutic interventions.

  • Inhibitors of RAS membrane interaction and subcellular localization remain attractive targets for therapeutic intervention. In this regard, recent discoveries of functionally relevant post-translational modifications of RAS, such as phosphorylation and ubiquitylation, suggest new opportunities to block RAS function.

  • RAS-driven cancers become efficient scavengers of nutrients and may rely on pathways such as autophagy and macropinocytosis. Furthermore, oncogenic KRAS has a crucial role in altering tumour metabolism, including rewiring of both glucose- and glutamine-dependent metabolic pathways. These alterations could provide the opportunity for novel therapeutic interventions.

Abstract

Despite more than three decades of intensive effort, no effective pharmacological inhibitors of the RAS oncoproteins have reached the clinic, prompting the widely held perception that RAS proteins are 'undruggable'. However, recent data from the laboratory and the clinic have renewed our hope for the development of RAS-inhibitory molecules. In this Review, we summarize the progress and the promise of five key approaches. Firstly, we focus on the prospects of using direct inhibitors of RAS. Secondly, we address the issue of whether blocking RAS membrane association is a viable approach. Thirdly, we assess the status of targeting RAS downstream effector signalling, which is arguably the most favourable current approach. Fourthly, we address whether the search for synthetic lethal interactors of mutant RAS still holds promise. Finally, RAS-mediated changes in cell metabolism have recently been described and we discuss whether these changes could be exploited for new therapeutic directions. We conclude with perspectives on how additional complexities, which are not yet fully understood, may affect each of these approaches.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Approaches to discover and develop pharmacological inhibitors of mutant RAS.
Figure 2: Compounds that have been reported to bind to RAS.
Figure 3: Inhibitors of RAS effector signalling under clinical evaluation.
Figure 4: Three-dimensional structures of RAS–ligand complexes.
Figure 5: Synthetic lethal interactors in RAS-mutant cancers.
Figure 6: RAS-driven alterations in metabolism.

Similar content being viewed by others

References

  1. Cox, A. D. & Der, C. J. RAS history: The saga continues. Small GTPases 1, 2–27 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Thompson, H. US National Cancer Institute's new RAS project targets an old foe. Nature Med. 19, 949–950 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. Hingorani, S. R. et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Ji, H. et al. LKB1 modulates lung cancer differentiation and metastasis. Nature 448, 807–810 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Haigis, K. M. et al. Differential effects of oncogenic KRAS and NRAS on proliferation, differentiation and tumor progression in the colon. Nature Genet. 40, 600–608 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Brummelkamp, T. R., Bernards, R. & Agami, R. Stable suppression of tumorigenicity by virus-mediated RNA interference. Cancer Cell 2, 243–247 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Lim, K. H. & Counter, C. M. Reduction in the requirement of oncogenic RAS signaling to activation of PI3K/AKT pathway during tumor maintenance. Cancer Cell 8, 381–392 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Singh, A. et al. A gene expression signature associated with “KRAS addiction” reveals regulators of EMT and tumor cell survival. Cancer Cell 15, 489–500 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chin, L. et al. Essential role for oncogenic RAS in tumour maintenance. Nature 400, 468–472 (1999).

    Article  CAS  PubMed  Google Scholar 

  12. Collins, M. A. et al. Metastatic pancreatic cancer is dependent on oncogenic KRAS in mice. PLoS ONE 7, e49707 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fisher, G. H. et al. Induction and apoptotic regression of lung adenocarcinomas by regulation of a KRAS transgene in the presence and absence of tumor suppressor genes. Genes Dev. 15, 3249–3262 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kwong, L. N. et al. Oncogenic NRAS signaling differentially regulates survival and proliferation in melanoma. Nature Med. 18, 1503–1510 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Ying, H. et al. Oncogenic KRAS maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell 149, 656–670 (2012). This study shows, in an autochthonous model, that oncogenic KRAS is required for pancreatic tumour maintenance, in part through its role in reprogramming anabolic glucose metabolism.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bos, J. L., Rehmann, H. & Wittinghofer, A. GEFs and GAPs: critical elements in the control of small G proteins. Cell 129, 865–877 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. 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 

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

    Article  CAS  Google Scholar 

  19. Reiss, Y., Goldstein, J. L., Seabra, M. C., Casey, P. J. & Brown, M. S. Inhibition of purified p21ras farnesyl:protein transferase by Cys-AAX tetrapeptides. Cell 62, 81–88 (1990).

    Article  CAS  PubMed  Google Scholar 

  20. James, G. L., Goldstein, J. L. & Brown, M. S. Polylysine and CVIM sequences of KRASB dictate specificity of prenylation and confer resistance to benzodiazepine peptidomimetic in vitro. J. Biol. Chem. 270, 6221–6226 (1995).

    Article  CAS  PubMed  Google Scholar 

  21. 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 

  22. Rowell, C. A., Kowalczyk, J. J., Lewis, M. D. & Garcia, A. M. Direct demonstration of geranylgeranylation and farnesylation of Ki-RAS in vivo. J. Biol. Chem. 272, 14093–14097 (1997).

    Article  CAS  PubMed  Google Scholar 

  23. Vigil, D., Cherfils, J., Rossman, K. L. & Der, C. J. RAS superfamily GEFs and GAPs: validated and tractable targets for cancer therapy? Nature Rev. Cancer 10, 842–857 (2010).

    Article  CAS  Google Scholar 

  24. 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 

  25. Buhrman, G. et al. Analysis of binding site hot spots on the surface of RAS GTPase. J. Mol. Biol. 413, 773–789 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. 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 

  28. Peri, F. et al. Design, synthesis and biological evaluation of sugar-derived RAS inhibitors. Chembiochem 6, 1839–1848 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  30. 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 

  31. Karaguni, I. M. et al. The new sulindac derivative IND 12 reverses RAS-induced cell transformation. Cancer Res. 62, 1718–1723 (2002).

    CAS  PubMed  Google Scholar 

  32. Karaguni, I. M. et al. New indene-derivatives with anti-proliferative properties. Bioorg. Med. Chem. Lett. 12, 709–713 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Gonzalez-Perez, 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  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kato-Stankiewicz, J. et al. Inhibitors of RAS/RAF1 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 

  35. 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 

  36. Patgiri, A., Yadav, K. K., Arora, P. S. & Bar-Sagi, D. An orthosteric inhibitor of the RAS–SOS interaction. Nature Chem. Biol. 7, 585–587 (2011).

    Article  CAS  Google Scholar 

  37. Chang, Y. S. et al. Stapled α-helical peptide drug development: a potent dual inhibitor of MDM2 and MDMX for p53-dependent cancer therapy. Proc. Natl Acad. Sci. USA 110, E3445–E3454 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Ostrem, J. M., Peters, U., Sos, M. L., Wells, J. A. & Shokat, K. M. KRAS (G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 503, 548–551 (2013). This study describes a novel approach to selectively target RAS-G12C by covalently attaching small molecules to the cysteine at residue 12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chen, K. X. et al. A novel class of highly potent irreversible hepatitis C virus NS5B polymerase inhibitors. J. Med. Chem. 55, 2089–2101 (2012).

    Article  CAS  PubMed  Google Scholar 

  43. Ward, R. A. et al. Structure- and reactivity-based development of covalent inhibitors of the activating and gatekeeper mutant forms of the epidermal growth factor receptor (EGFR). J. Med. Chem. 56, 7025–7048 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Lim, S. M. et al. Therapeutic targeting of oncogenic KRAS by a covalent catalytic site inhibitor. Angew. Chem. Int. Ed Engl. 53, 199–204 (2014). This study describes an approach to selectively target G12C mutant RAS by covalently attaching a nucleotide analogue.

    Article  CAS  PubMed  Google Scholar 

  45. 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). In this paper, compounds were described that activate SOS-mediated nucleotide exchange and paradoxically inhibit downstream RAS signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen, X., Makarewicz, J. M., Knauf, J. A., Johnson, L. K. & Fagin, J. A. Transformation by HrasG12V is consistently associated with mutant allele copy gains and is reversed by farnesyl transferase inhibition. Oncogene http://dx.doi.org/10.1038/onc.2013.489 (2013).

  47. Liu, M. et al. Targeting the protein prenyltransferases efficiently reduces tumor development in mice with KRAS-induced lung cancer. Proc. Natl Acad. Sci. USA 107, 6471–6476 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Marom, M. et al. Selective inhibition of RAS-dependent cell growth by farnesylthiosalisylic acid. J. Biol. Chem. 270, 22263–22270 (1995).

    Article  CAS  PubMed  Google Scholar 

  49. Gana-Weisz, M. et al. The RAS antagonist S-farnesylthiosalicylic acid induces inhibition of MAPK activation. Biochem. Biophys. Res. Commun. 239, 900–904 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Haklai, R. et al. Dislodgment and accelerated degradation of RAS. Biochemistry 37, 1306–1314 (1998).

    Article  CAS  PubMed  Google Scholar 

  51. Makovski, V., Haklai, R. & Kloog, Y. Farnesylthiosalicylic acid (salirasib) inhibits RHEB in TSC2-null ELT3 cells: a potential treatment for lymphangioleiomyomatosis. Int. J. Cancer 130, 1420–1429 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. McMahon, L. P., Yue, W., Santen, R. J. & Lawrence, J. C. Jr. Farnesylthiosalicylic acid inhibits mammalian target of rapamycin (mTOR) activity both in cells and in vitro by promoting dissociation of the mTOR-Raptor complex. Mol. Endocrinol. 19, 175–183 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Hanker, A. B. et al. Differential requirement of CAAX-mediated posttranslational processing for RHEB localization and signaling. Oncogene 29, 380–391 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Weisz, B. et al. A new functional RAS antagonist inhibits human pancreatic tumor growth in nude mice. Oncogene 18, 2579–2588 (1999).

    Article  CAS  PubMed  Google Scholar 

  55. Haklai, R., Elad-Sfadia, G., Egozi, Y. & Kloog, Y. Orally administered FTS (salirasib) inhibits human pancreatic tumor growth in nude mice. Cancer Chemother. Pharmacol. 61, 89–96 (2008).

    Article  CAS  PubMed  Google Scholar 

  56. 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 

  57. Wahlstrom, A. M. et al. Rce1 deficiency accelerates the development of KRAS-induced myeloproliferative disease. Blood 109, 763–768 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Court, H. et al. Isoprenylcysteine carboxylmethyltransferase deficiency exacerbates KRAS-driven pancreatic neoplasia via Notch suppression. J. Clin. Invest. 123, 4681–4694 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Majmudar, J. D. et al. Amide-modified prenylcysteine based ICMT inhibitors: structure–activity relationships, kinetic analysis and cellular characterization. Bioorg. Med. Chem. 20, 283–295 (2012).

    Article  CAS  PubMed  Google Scholar 

  61. Manandhar, S. P., Hildebrandt, E. R. & Schmidt, W. K. Small-molecule inhibitors of the RCE1P CAAX protease. J. Biomol. Screen 12, 983–993 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Winter-Vann, A. M. et al. A small-molecule inhibitor of isoprenylcysteine carboxyl methyltransferase with antitumor activity in cancer cells. Proc. Natl Acad. Sci. USA 102, 4336–4341 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chiu, V. K. et al. RAS signalling on the endoplasmic reticulum and the Golgi. Nature Cell Biol. 4, 343–350 (2002).

    Article  CAS  PubMed  Google Scholar 

  64. Cuiffo, B. & Ren, R. Palmitoylation of oncogenic NRAS is essential for leukemogenesis. Blood 115, 3598–3605 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Swarthout, J. T. et al. DHHC9 and GCP16 constitute a human protein fatty acyltransferase with specificity for H- and N-RAS. J. Biol. Chem. 280, 31141–31148 (2005).

    Article  CAS  PubMed  Google Scholar 

  66. Goodwin, J. S. et al. Depalmitoylated RAS traffics to and from the Golgi complex via a nonvesicular pathway. J. Cell Biol. 170, 261–272 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. 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 

  68. 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 

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

    Article  CAS  Google Scholar 

  70. Resh, M. D. Targeting protein lipidation in disease. Trends Mol. Med. 18, 206–214 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Bivona, T. G. et al. PKC regulates a farnesyl-electrostatic switch on KRAS that promotes its association with BCL-XL on mitochondria and induces apoptosis. Mol. Cell 21, 481–493 (2006). This study identified a kinase-driven phosphorylation that regulates KRAS4B membrane association, subcellular localization and oncogenic properties.

    Article  CAS  PubMed  Google Scholar 

  72. Sung, P. J. et al. Phosphorylated KRAS limits cell survival by blocking BCL-XL sensitization of inositol trisphosphate receptors. Proc. Natl Acad. Sci. USA 110, 20593–20598 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kollar, P., Rajchard, J., Balounova, Z. & Pazourek, J. Marine natural products: bryostatins in preclinical and clinical studies. Pharm. Biol. 52, 237–242 (2013).

    Article  CAS  PubMed  Google Scholar 

  74. Barcelo, C. et al. Phosphorylation at Ser181 of oncogenic KRAS is required for tumor growth. Cancer Res. 74, 1190–1199 (2013).

    Article  CAS  PubMed  Google Scholar 

  75. Lim, K. H., Ancrile, B. B., Kashatus, D. F. & Counter, C. M. Tumour maintenance is mediated by eNOS. Nature 452, 646–649 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lampson, B. L. et al. Targeting eNOS in pancreatic cancer. Cancer Res. 72, 4472–4482 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. 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 

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

    Article  CAS  Google Scholar 

  79. Zimmermann, G. et al. Small molecule inhibition of the KRAS–PDEδ interaction impairs oncogenic KRAS signalling. Nature 497, 638–642 (2013). This study identified a small molecule that can disrupt KRAS4B association with a protein chaperone, altering KRAS4B membrane trafficking and cellular activity.

    Article  CAS  PubMed  Google Scholar 

  80. Philips, M. R. RAS hitchhikes on PDE6δ. Nature Cell Biol. 14, 128–129 (2012).

    Article  CAS  PubMed  Google Scholar 

  81. Jura, N., Scotto-Lavino, E., Sobczyk, A. & Bar-Sagi, D. Differential modification of RAS proteins by ubiquitination. Mol. Cell 21, 679–687 (2006).

    Article  CAS  PubMed  Google Scholar 

  82. Sasaki, A. T. et al. Ubiquitination of KRAS enhances activation and facilitates binding to select downstream effectors. Sci. Signal 4, ra13 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Sumita, K. et al. Degradation of activated KRAS orthologue via KRAS-specific lysine residues is required for cytokinesis. J. Biol. Chem. 289, 3950–3959 (2014).

    Article  CAS  PubMed  Google Scholar 

  84. Yang, M. H. et al. HDAC6 and SIRT2 regulate the acetylation state and oncogenic activity of mutant KRAS. Mol. Cancer Res. 11, 1072–1077 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. 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 

  86. Blasco, R. B. et al. CRAF, but not BRAF, is essential for development of KRAS oncogene-driven non-small cell lung carcinoma. Cancer Cell 19, 652–663 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Collisson, E. A. et al. A central role for RAF→MEK→ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2, 685–693 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Roskoski, R. Jr. ERK1/2 MAP kinases: structure, function, and regulation. Pharmacol. Res. 66, 105–143 (2012).

    Article  CAS  PubMed  Google Scholar 

  89. Lyons, J. F., Wilhelm, S., Hibner, B. & Bollag, G. Discovery of a novel RAF kinase inhibitor. Endocr. Relat. Cancer 8, 219–225 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Wilhelm, S. M. et al. 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, 7099–7109 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  92. Hatzivassiliou, G. et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 464, 431–435 (2010).

    Article  CAS  PubMed  Google Scholar 

  93. Heidorn, S. J. et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 140, 209–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Poulikakos, P. I., Zhang, C., Bollag, G., Shokat, K. M. & Rosen, N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature 464, 427–430 (2010). References 92–94 determined that BRAF inhibitors cause RAS-activation-dependent RAF dimerization and ERK activation rather than inactivation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Hall-Jackson, C. A. et al. Paradoxical activation of RAF by a novel RAF inhibitor. Chem. Biol. 6, 559–568 (1999).

    Article  CAS  PubMed  Google Scholar 

  96. Oberholzer, P. A. et al. RAS mutations are associated with the development of cutaneous squamous cell tumors in patients treated with RAF inhibitors. J. Clin. Oncol. 30, 316–321 (2012).

    Article  CAS  PubMed  Google Scholar 

  97. Su, F. et al. RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors. N. Engl. J. Med. 366, 207–215 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Freeman, A. K., Ritt, D. A. & Morrison, D. K. Effects of RAF dimerization and its inhibition on normal and disease-associated RAF signaling. Mol. Cell 49, 751–758 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Gilmartin, A. G. et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin. Cancer Res. 17, 989–1000 (2011).

    Article  CAS  PubMed  Google Scholar 

  100. Engelman, J. A. et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nature Med. 14, 1351–1356 (2008).

    Article  CAS  PubMed  Google Scholar 

  101. Hatzivassiliou, G. et al. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature 501, 232–236 (2013).

    Article  CAS  PubMed  Google Scholar 

  102. Ishii, N. et al. Enhanced inhibition of ERK signaling by a novel allosteric MEK inhibitor, CH5126766, that suppresses feedback reactivation of RAF activity. Cancer Res. 73, 4050–4060 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Duncan, J. S. et al. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell 149, 307–321 (2012). This study applied a kinome-wide approach to identify MEK-inhibitor-induced compensatory events that can then be targeted to prevent de novo resistance mechanisms.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Little, A. S. et al. Amplification of the driving oncogene, KRAS or BRAF, underpins acquired resistance to MEK1/2 inhibitors in colorectal cancer cells. Sci. Signal. 4, ra17 (2011).

    Article  CAS  PubMed  Google Scholar 

  105. Bollag, G. et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature 467, 596–599 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Morris, E. J. et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 3, 742–750 (2013).

    Article  CAS  PubMed  Google Scholar 

  107. Gupta, S. et al. Binding of RAS to phosphoinositide 3-kinase p110 α is required for RAS-driven tumorigenesis in mice. Cell 129, 957–968 (2007).

    Article  CAS  PubMed  Google Scholar 

  108. Castellano, E. et al. Requirement for interaction of PI3-kinase p110α with RAS in lung tumor maintenance. Cancer Cell 24, 617–630 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Ebi, H. et al. Receptor tyrosine kinases exert dominant control over PI3K signaling in human KRAS mutant colorectal cancers. J. Clin. Invest. 121, 4311–4321 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Britten, C. D. PI3K and MEK inhibitor combinations: examining the evidence in selected tumor types. Cancer Chemother. Pharmacol. 71, 1395–1409 (2013).

    Article  CAS  PubMed  Google Scholar 

  111. Bodemann, B. O. & White, M. A. RAL GTPases and cancer: linchpin support of the tumorigenic platform. Nature Rev. Cancer 8, 133–140 (2008).

    Article  CAS  Google Scholar 

  112. Neel, N. F. et al. The RALGEF–RAL effector signaling network: the road less traveled for anti-RAS drug discovery. Genes Cancer 2, 275–287 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Gonzalez-Garcia, A. et al. RALGDS is required for tumor formation in a model of skin carcinogenesis. Cancer Cell 7, 219–226 (2005).

    Article  CAS  PubMed  Google Scholar 

  114. Vigil, D. et al. Aberrant overexpression of the RGL2 RAL small GTPase-specific guanine nucleotide exchange factor promotes pancreatic cancer growth through RAL-dependent and RAL-independent mechanisms. J. Biol. Chem. 285, 34729–34740 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Lim, K. H. et al. Divergent roles for RALA and RALB in malignant growth of human pancreatic carcinoma cells. Curr. Biol. 16, 2385–2394 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Peschard, P. et al. Genetic deletion of RALA and RALB small GTPases reveals redundant functions in development and tumorigenesis. Curr. Biol. 22, 2063–2068 (2012).

    Article  CAS  PubMed  Google Scholar 

  117. Wu, J. C. et al. Identification of V23RALA-Ser194 as a critical mediator for Aurora-A-induced cellular motility and transformation by small pool expression screening. J. Biol. Chem. 280, 9013–9022 (2005).

    Article  CAS  PubMed  Google Scholar 

  118. Lim, K. H. et al. Aurora-A phosphorylates, activates, and relocalizes the small GTPase RalA. Mol. Cell. Biol. 30, 508–523 (2010).

    Article  CAS  PubMed  Google Scholar 

  119. Wang, H. et al. Phosphorylation of RALB is important for bladder cancer cell growth and metastasis. Cancer Res. 70, 8760–8769 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Martin, T. D., Mitin, N., Cox, A. D., Yeh, J. J. & Der, C. J. Phosphorylation by protein kinase Cα regulates RALB small GTPase protein activation, subcellular localization, and effector utilization. J. Biol. Chem. 287, 14827–14836 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Chien, Y. et al. RALB GTPase-mediated activation of the IκB family kinase TBK1 couples innate immune signaling to tumor cell survival. Cell 127, 157–170 (2006).

    Article  CAS  PubMed  Google Scholar 

  122. Martin, T. D. et al. RAL and RHEB GTPase activating proteins integrate mTOR and GTPase signaling in aging, autophagy, and tumor cell invasion. Mol. Cell 53, 209–220 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Malliri, A. et al. Mice deficient in the RAC activator TIAM1 are resistant to RAS-induced skin tumours. Nature 417, 867–871 (2002).

    Article  CAS  PubMed  Google Scholar 

  124. Lambert, J. M. et al. TIAM1 mediates RAS activation of RAC by a PI(3)K-independent mechanism. Nature Cell Biol. 4, 621–625 (2002).

    Article  CAS  PubMed  Google Scholar 

  125. Welch, H. C. et al. P-REX1, a PtdIns(3,4,5)P3- and Gβγ-regulated guanine-nucleotide exchange factor for RAC. Cell 108, 809–821 (2002).

    Article  CAS  PubMed  Google Scholar 

  126. Hodis, E. et al. A landscape of driver mutations in melanoma. Cell 150, 251–263 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Krauthammer, M. et al. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nature Genet. 44, 1006–1014 (2012).

    Article  CAS  PubMed  Google Scholar 

  128. Kissil, J. L. et al. Requirement for RAC1 in a KRAS induced lung cancer in the mouse. Cancer Res. 67, 8089–8094 (2007).

    Article  CAS  PubMed  Google Scholar 

  129. Heid, I. et al. Early requirement of RAC1 in a mouse model of pancreatic cancer. Gastroenterol 141, 719–730.e7 (2011).

    Article  CAS  Google Scholar 

  130. Cardama, G. A. et al. Preclinical development of novel RAC1–GEF signaling inhibitors using a rational design approach in highly aggressive breast cancer cell lines. Anticancer Agents Med. Chem. 14, 840–851 (2013).

    Article  CAS  Google Scholar 

  131. Gao, Y., Dickerson, J. B., Guo, F., Zheng, J. & Zheng, Y. Rational design and characterization of a RAC GTPase-specific small molecule inhibitor. Proc. Natl Acad. Sci. USA 101, 7618–7623 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Chow, H. Y. et al. p21-activated kinase 1 is required for efficient tumor formation and progression in a RAS-mediated skin cancer model. Cancer Res. 72, 5966–5975 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Fritsch, R. et al. RAS and RHO families of GTPases directly regulate distinct phosphoinositide 3-kinase isoforms. Cell 153, 1050–1063 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Chan, J. J. & Katan, M. PLCε and the RASSF family in tumour suppression and other functions. Adv. Biol. Regul. 53, 258–279 (2013).

    Article  CAS  PubMed  Google Scholar 

  135. Hartman, J. L. 4th, Garvik, B. & Hartwell, L. Principles for the buffering of genetic variation. Science 291, 1001–1004 (2001).

    Article  CAS  PubMed  Google Scholar 

  136. Kaelin, W. G. Jr. The concept of synthetic lethality in the context of anticancer therapy. Nature Rev. Cancer 5, 689–698 (2005).

    Article  CAS  Google Scholar 

  137. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).

    Article  CAS  PubMed  Google Scholar 

  138. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  139. Luo, J., Solimini, N. L. & Elledge, S. J. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136, 823–837 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Yu, B. & Luo, J. in Inhibitors of the RAS Superfamily G-proteins, Part B. (eds. Tamanoi, F. & Der, C. J.) 201–215 (Academic Press, 2013).

    Book  Google Scholar 

  141. Cox, A. D. & Der, C. J. The dark side of RAS: regulation of apoptosis. Oncogene 22, 8999–9006 (2003).

    Article  CAS  PubMed  Google Scholar 

  142. Kumar, M. S. et al. The GATA2 transcriptional network is requisite for RAS oncogene-driven non-small cell lung cancer. Cell 149 642–655 (2012).

    Article  CAS  PubMed  Google Scholar 

  143. Luo, J. et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the RAS oncogene. Cell 137, 835–848 (2009). This is one of several studies describing large-scale RNAi screens aiming to identify synthetic lethal partners of the KRAS oncogene.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Barbie, D. A. et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Steckel, M. et al. Determination of synthetic lethal interactions in KRAS oncogene-dependent cancer cells reveals novel therapeutic targeting strategies. Cell Res. 22, 1227–1245 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Carretero, J. et al. Integrative genomic and proteomic analyses identify targets for LKB1-deficient metastatic lung tumors. Cancer Cell 17, 547–559 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Kim, H. S. et al. Systematic identification of molecular subtype-selective vulnerabilities in non-small-cell lung cancer. Cell 155, 552–566 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  149. Kapoor, A. et al. YAP1 activation enables bypass of oncogenic KRAS addiction in pancreatic cancer. Cell 158, 185–197 (2014). This study, along with reference 150, identified a novel bypass mechanism that could render KRAS-addicted cancer cells resistant to RAS-targeted therapy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Shao, D. D. et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 158, 171–184 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Cheung, H. W. et al. Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer. Proc. Natl Acad. Sci. USA 108, 12372–12377 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Marcotte, R. et al. Essential gene profiles in breast, pancreatic, and ovarian cancer cells. Cancer Discov. 2, 172–189 (2012).

    Article  CAS  PubMed  Google Scholar 

  153. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Anderson, S. N., Towne, D. L., Burns, D. J. & Warrior, U. A high-throughput soft agar assay for identification of anticancer compound. J. Biomol. Screen 12, 938–945 (2007).

    Article  CAS  PubMed  Google Scholar 

  155. Meacham, C. E., Ho, E. E., Dubrovsky, E., Gertler, F. B. & Hemann, M. T. In vivo RNAi screening identifies regulators of actin dynamics as key determinants of lymphoma progression. Nature Genet. 41, 1133–1137 (2009).

    Article  CAS  PubMed  Google Scholar 

  156. Zuber, J. et al. An integrated approach to dissecting oncogene addiction implicates a MYB-coordinated self-renewal program as essential for leukemia maintenance. Genes Dev. 25, 1628–1640 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Possemato, R. et al. Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature 476, 346–350 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Murugaesu, N. et al. An in vivo functional screen identifies ST6GalNAc2 sialyltransferase as a breast cancer metastasis suppressor. Cancer Discov. 4, 304–317 (2014).

    Article  CAS  PubMed  Google Scholar 

  159. Schramek, D. et al. Direct in vivo RNAi screen unveils myosin IIa as a tumor suppressor of squamous cell carcinomas. Science 343, 309–313 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Zender, L. et al. An oncogenomics-based in vivo RNAi screen identifies tumor suppressors in liver cancer. Cell 135, 852–864 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Kumar, M. S. et al. The GATA2 transcriptional network is requisite for RAS oncogene-driven non-small cell lung cancer. Cell 149, 642–655 (2012).

    Article  CAS  PubMed  Google Scholar 

  162. Luo, T. et al. STK33 kinase inhibitor BRD-8899 has no effect on KRAS-dependent cancer cell viability. Proc. Natl Acad. Sci. USA 109, 2860–2865 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Muvaffak, A. et al. Evaluating TBK1 as a therapeutic target in cancers with activated IRF3. Mol. Cancer Res. 12, 1055–1066 (2014).

    Article  CAS  PubMed  Google Scholar 

  164. Zhu, Z. et al. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov. 4, 452–465 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Corcoran, R. B. et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. Cancer Cell 23, 121–128 (2013).

    Article  CAS  PubMed  Google Scholar 

  166. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nature Protoc. 8, 2281–2308 (2013).

    Article  CAS  Google Scholar 

  167. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  168. Wang, T., Wei, J. J., Sabatini, D. M. & Lander, E. S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014).

    Article  CAS  PubMed  Google Scholar 

  169. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. DeBerardinis, R. J. & Thompson, C. B. Cellular metabolism and disease: what do metabolic outliers teach us? Cell 148, 1132–1144 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Lunt, S. Y. & Vander Heiden, M. G. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 27, 441–464 (2011).

    Article  CAS  PubMed  Google Scholar 

  172. Stine, Z. E. & Dang, C. V. Stress eating and tuning out: cancer cells re-wire metabolism to counter stress. Crit. Rev. Biochem. Mol. Biol. 48, 609–619 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Levine, A. J. & Puzio-Kuter, A. M. The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 330, 1340–1344 (2010).

    Article  CAS  PubMed  Google Scholar 

  174. Racker, E., Resnick, R. J. & Feldman, R. Glycolysis and methylaminoisobutyrate uptake in rat-1 cells transfected with ras or myc oncogenes. Proc. Natl Acad. Sci. USA 82, 3535–3538 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Wise, D. R. et al. MYC regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc. Natl Acad. Sci. USA 105, 18782–18787 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Son, J. et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 496, 101–105 (2013). This paper shows that pancreatic cancers depend on glutamine, which is utilized by a KRAS-regulated pathway that is crucial for maintaining redox homeostasis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. White, E. Exploiting the bad eating habits of RAS-driven cancers. Genes Dev. 27, 2065–2071 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Yang, Z. & Klionsky, D. J. Eaten alive: a history of macroautophagy. Nature Cell Biol. 12, 814–822 (2010).

    Article  CAS  PubMed  Google Scholar 

  179. Kimmelman, A. C. The dynamic nature of autophagy in cancer. Genes Dev. 25, 1999–2010 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Mah, L. Y. & Ryan, K. M. Autophagy and cancer. Cold Spring Harb. Perspect. Biol. 4, a008821 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  181. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nature Rev. Cancer 12, 401–410 (2012).

    Article  CAS  Google Scholar 

  182. Guo, J. Y. et al. Activated RAS requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 25, 460–470 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Guo, J. Y. et al. Autophagy suppresses progression of KRAS-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev. 27, 1447–1461 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Lock, R. et al. Autophagy facilitates glycolysis during RAS-mediated oncogenic transformation. Mol. Biol. Cell 22, 165–178 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Yang, S. et al. Pancreatic cancers require autophagy for tumor growth. Genes Dev. 25, 717–729 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Rao, S. et al. A dual role for autophagy in a murine model of lung cancer. Nature Commun. 5, 3056 (2014).

    Article  CAS  Google Scholar 

  187. Rosenfeldt, M. T. et al. p53 status determines the role of autophagy in pancreatic tumour development. Nature 504, 296–300 (2013).

    Article  CAS  PubMed  Google Scholar 

  188. Yang, A. et al. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov. 4, 905–913 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Amaravadi, R. K. et al. Principles and current strategies for targeting autophagy for cancer treatment. Clin. Cancer Res. 17, 654–666 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Mancias, J. D. & Kimmelman, A. C. Targeting autophagy addiction in cancer. Oncotarget 2, 1302–1306 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  191. 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 

  192. Yang, Z. & Klionsky, D. J. Mammalian autophagy: core molecular machinery and signaling regulation. Curr. Opin. Cell Biol. 22, 124–131 (2010).

    Article  CAS  PubMed  Google Scholar 

  193. Bar-Sagi, D. & Feramisco, J. R. Induction of membrane ruffling and fluid-phase pinocytosis in quiescent fibroblasts by ras proteins. Science 233, 1061–1068 (1986).

    Article  CAS  PubMed  Google Scholar 

  194. Lim, J. P. & Gleeson, P. A. Macropinocytosis: an endocytic pathway for internalising large gulps. Immunol. Cell Biol. 89, 836–843 (2011).

    Article  CAS  PubMed  Google Scholar 

  195. Commisso, C. et al. Macropinocytosis of protein is an amino acid supply route in RAS-transformed cells. Nature 497, 633–637 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Kamphorst, J. J. et al. Hypoxic and RAS-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc. Natl Acad. Sci. USA 110, 8882–8887 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Yun, J. et al. Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells. Science 325, 1555–1559 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. McCleland, M. L. et al. Lactate dehydrogenase B is required for the growth of KRAS-dependent lung adenocarcinomas. Clin. Cancer Res. 19, 773–784 (2013).

    Article  CAS  PubMed  Google Scholar 

  199. Patra, K. C. et al. Hexokinase 2 is required for tumor initiation and maintenance and its systemic deletion is therapeutic in mouse models of cancer. Cancer Cell 24, 213–228 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Gaglio, D. et al. Oncogenic KRAS decouples glucose and glutamine metabolism to support cancer cell growth. Mol. Syst. Biol. 7, 523 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Weinberg, F. et al. Mitochondrial metabolism and ROS generation are essential for KRAS-mediated tumorigenicity. Proc. Natl Acad. Sci. USA 107, 8788–8793 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Shukla, K. et al. Design, synthesis, and pharmacological evaluation of bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide 3 (BPTES) analogs as glutaminase inhibitors. J. Med. Chem. 55, 10551–10563 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Thornburg, J. M. et al. Targeting aspartate aminotransferase in breast cancer. Breast Cancer Res. 10, R84 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Nguyen, T., Nioi, P. & Pickett, C. B. The NRF2-antioxidant response element signaling pathway and its activation by oxidative stress. J. Biol. Chem. 284, 13291–13295 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. DeNicola, G. M. et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 475, 106–109 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. To, M. D. et al. Kras regulatory elements and exon 4A determine mutation specificity in lung cancer. Nature Genet. 40, 1240–1244 (2008).

    Article  CAS  PubMed  Google Scholar 

  207. Lampson, B. L. et al. Rare codons regulate KRAS oncogenesis. Curr. Biol. 23, 70–75 (2013).

    Article  CAS  PubMed  Google Scholar 

  208. Abubaker, J. et al. Prognostic significance of alterations in KRAS isoforms KRAS4A/4B and KRAS mutations in colorectal carcinoma. J. Pathol. 219, 435–445 (2009).

    Article  CAS  PubMed  Google Scholar 

  209. De Roock, W. et al. Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA 304, 1812–1820 (2010).

    Article  CAS  PubMed  Google Scholar 

  210. Tejpar, S. et al. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J. Clin. Oncol. 30, 3570–3577 (2012).

    Article  CAS  PubMed  Google Scholar 

  211. Ihle, N. T. et al. Effect of KRAS oncogene substitutions on protein behavior: implications for signaling and clinical outcome. J. Natl Cancer Inst. 104, 228–239 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Zhang, Z. et al. Wildtype Kras2 can inhibit lung carcinogenesis in mice. Nature Genet. 29, 25–33 (2001).

    Article  CAS  PubMed  Google Scholar 

  213. Bremner, R. & Balmain, A. Genetic changes in skin tumor progression: correlation between presence of a mutant ras gene and loss of heterozygosity on mouse chromosome 7. Cell 61, 407–417 (1990).

    Article  CAS  PubMed  Google Scholar 

  214. Li, H. et al. Growth inhibitory effect of wild-type KRAS2 gene on a colonic adenocarcinoma cell line. World J. Gastroenterol. 13, 934–938 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Qiu, W. et al. Disruption of p16 and activation of KRAS in pancreas increase ductal adenocarcinoma formation and metastasis in vivo. Oncotarget 2, 862–873 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  216. Lin, W. C. et al. HRAS forms dimers on membrane surfaces via a protein–protein interface. Proc. Natl Acad. Sci. USA 111, 2996–3001 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Xu, J. et al. Dominant role of oncogene dosage and absence of tumor suppressor activity in NRAS-driven hematopoietic transformation. Cancer Discov. 3, 993–1001 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. 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 

  220. Vonderheide, R. H. & Nathanson, K. L. Immunotherapy at large: the road to personalized cancer vaccines. Nature Med. 19, 1098–1100 (2013).

    Article  CAS  PubMed  Google Scholar 

  221. Bayne, L. J. et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 21, 822–835 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Pylayeva-Gupta, Y., Lee, K. E., Hajdu, C. H., Miller, G. & Bar-Sagi, D. Oncogenic KRAS-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 21, 836–847 (2012). This study, along with reference 221, defined crucial mechanisms of crosstalk between the immune system and tumour development.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Pecot, C. V., Calin, G. A., Coleman, R. L., Lopez-Berestein, G. & Sood, A. K. RNA interference in the clinic: challenges and future directions. Nature Rev. Cancer 11, 59–67 (2011).

    Article  CAS  Google Scholar 

  224. Thompson, H. J. et al. Sulfone metabolite of sulindac inhibits mammary carcinogenesis. Cancer Res. 57, 267–271 (1997).

    CAS  PubMed  Google Scholar 

  225. Sarthy, A. V. et al. Survivin depletion preferentially reduces the survival of activated KRAS-transformed cells. Mol. Cancer Ther. 6, 269–276 (2007).

    Article  CAS  PubMed  Google Scholar 

  226. Morgan-Lappe, S. E. et al. Identification of RAS-related nuclear protein, targeting protein for Xenopus kinesin-like protein 2, and stearoyl-CoA desaturase 1 as promising cancer targets from an RNAi-based screen. Cancer Res. 67, 4390–4398 (2007).

    Article  CAS  PubMed  Google Scholar 

  227. Scholl, C. et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell 137, 821–834 (2009).

    Article  CAS  PubMed  Google Scholar 

  228. Vicent, S. et al. Wilms tumor 1 (WT1) regulates KRAS-driven oncogenesis and senescence in mouse and human models. J. Clin. Invest. 120, 3940–3952 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Wang, Y. et al. Critical role for transcriptional repressor SNAIL2 in transformation by oncogenic RAS in colorectal carcinoma cells. Oncogene 29, 4658–4670 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Singh, A. et al. TAK1 inhibition promotes apoptosis in KRAS-dependent colon cancers. Cell 148, 639–650 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Cullis, J. et al. The RHOGEF GEF-H1 is required for oncogenic RAS signaling via KSR1. Cancer Cell 25, 181–195 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The work of C.J.D. and A.D.C. is supported by US National Institutes of Health (NIH) grants CA042978, CA179193 and CA175747, and by grants from the Lustgarten Foundation for Pancreatic Cancer Research, USA, and the Pancreatic Cancer Action Network-American Association for Cancer Research. The work of S.W.F. is supported by NIH grants DP1OD006933/DP1CA174419 (NIH Director's Pioneer Award; S.W.F.), P50A095103-12 (NCI SPORE in GI Cancer; R. J. Coffey), and RC2CA148375 (NIH ARRA Stimulus Grant; L. J. Marnett), and by the Lustgarten Foundation for Pancreatic Cancer Research. The work of A.C.K. is supported by NIH grant R01CA157490, American Cancer Society Research Scholar Grant RSG-13-298-01-TBG and by the Lustgarten Foundation. A.C.K. is a consultant for Forma Therapeutics. The work of J.L. is supported by the US National Cancer Institute Intramural Program.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Channing J. Der.

Ethics declarations

Competing interests

C.J.D. has received honoraria from Novartis, AstraZeneca and Eli Lilly and grant support from GlaxoSmithKline and Onconova. A.C.K. is a consultant for Forma Therapeutics. A.D.C., S.W.F. and J.L. declare no competing interests.

Related links

PowerPoint slides

Supplementary information

Supplementary information S1 (table)

Cancer gene mutation frequencies (PDF 89 kb)

Supplementary information S2 (table)

Frequency of RAS mutations in human cancers (PDF 531 kb)

Supplementary information S3 (figure)

RAS mutations in human cancer (PDF 153 kb)

Glossary

Undruggable

Druggable biological targets such as some proteins can be bound with high affinity by small molecules, natural products, or antibodies in such a way that the binding then alters target function to achieve a therapeutic benefit. Undruggable targets are not amenable to such interventions.

RNA interference

(RNAi). A method of gene silencing in which short, double-stranded RNA molecules degrade target mRNAs in a sequence-specific manner to inhibit the expression and function of genes of interest.

GTPase

An enzyme that binds and hydrolyzes GTP to GDP. GTP binding and hydrolysis take place within a highly conserved G-domain shared among all GTPases.

Isoprenoid

A class of organic lipid compounds made up of two or more structural units derived from isoprene, a five-carbon hydrocarbon with a branched-chain structure.

Prenylation

The covalent and irreversible addition of hydrophobic 15- or 20-carbon isoprenyl groups to the carboxy-terminus of proteins with an appropriate signal motif. This modification facilitates protein attachment to cell membranes.

NMR

NMR (nuclear magnetic resonance) is the selective absorption of electromagnetic radiation by an atomic nucleus in the presence of a strong, static magnetic field. NMR spectroscopy is often used to determine the structure and dynamics of proteins in solution.

Fragment-based screening

A method in drug discovery used to find lead compounds, based on initial identification of small chemical fragments that may bind only weakly to the biological target. By combining the fragments, a higher affinity lead compound can be designed.

Electrophile

A reagent attracted to electrons that participates in a chemical reaction by accepting an electron pair in order to bond to a nucleophile. A nucleophile is a reactant that provides a pair of electrons to form a new covalent bond.

Driver

Driver mutations confer a growth and/or survival advantage on cancer cells; they may or may not be required for maintenance of advanced malignancies. This contrasts with passenger mutations, which do not confer such an advantage and therefore do not contribute to cancer development.

Allosteric regulation

Regulation of an enzyme by binding of a molecule at a site that is not directly involved in its enzymatic activity. This binding often results in a conformational change that then enhances or decreases enzyme activity.

Achilles' heel

A deadly weakness or vulnerable point whose targeting can lead to the downfall of an otherwise strong entity (e.g., protein).

Synthetic lethality

A genetic interaction whereby two otherwise non-lethal genetic mutations lead to cell death.

CRISPR

CRISPR (clustered regularly interspaced short palindromic repeat) is an RNA-guided gene-editing platform that utilizes the bacterial protein Cas9 and a synthetic guide RNA to introduce a double-stranded DNA break at a specific location within the genome.

Autophagy

A cellular mechanism of 'self-eating' in which unneeded or dysfunctional cellular components are degraded by the lysosome and made available for recycling. Autophagy can assist in the stress response and support cellular survival by maintaining cellular energy levels during nutrient deficiency.

Macropinocytosis

An actin-driven mechanism of endocytosis in which large fluid droplets are trapped in a large organelle originating from plasma membrane extensions (ruffles) of the cell surface.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cox, A., Fesik, S., Kimmelman, A. et al. Drugging the undruggable RAS: Mission Possible?. Nat Rev Drug Discov 13, 828–851 (2014). https://doi.org/10.1038/nrd4389

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd4389

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