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.

  • Original Article
  • Published:

mTORC1 upregulation via ERK-dependent gene expression change confers intrinsic resistance to MEK inhibitors in oncogenic KRas-mutant cancer cells

Abstract

Cancer cells harboring oncogenic BRaf mutants, but not oncogenic KRas mutants, are sensitive to MEK inhibitors (MEKi). The mechanism underlying the intrinsic resistance to MEKi in KRas-mutant cells is under intensive investigation. Here, we pursued this mechanism by live imaging of extracellular signal-regulated kinases (ERK) and mammalian target of rapamycin complex 1 (mTORC1) activities in oncogenic KRas or BRaf-mutant cancer cells. We established eight cancer cell lines expressing Förster resonance energy transfer (FRET) biosensors for ERK activity and S6K activity, which was used as a surrogate marker for mTORC1 activity. Under increasing concentrations of MEKi, ERK activity correlated linearly with the cell growth rate in BRaf-mutant cancer cells, but not KRas-mutant cancer cells. The administration of PI3K inhibitors resulted in a linear correlation between ERK activity and cell growth rate in KRas-mutant cancer cells. Intriguingly, mTORC1 activity was correlated linearly with the cell growth rate in both BRaf-mutant cancer cells and KRas-mutant cancer cells. These observations suggested that mTORC1 activity had a pivotal role in cell growth and that the mTORC1 activity was maintained primarily by the ERK pathway in BRaf-mutant cancer cells and by both the ERK and PI3K pathways in KRas-mutant cancer cells. FRET imaging revealed that MEKi inhibited mTORC1 activity with slow kinetics, implying transcriptional control of mTORC1 activity by ERK. In agreement with this observation, MEKi induced the expression of negative regulators of mTORC1, including TSC1, TSC2 and Deptor, which occurred more significantly in BRaf-mutant cells than in KRas-mutant cells. These findings suggested that the suppression of mTORC1 activity and induction of negative regulators of mTORC1 in cancer cells treated for at least 1 day could be used as surrogate markers for the MEKi sensitivity of cancer cells.

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
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

References

  1. Nishida E, Gotoh Y . The MAP kinase cascade is essential for diverse signal transduction pathways. Trends Biochem Sci 1993; 18: 128–131.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Weinstein IB . Cancer. Addiction to oncogenes—the Achilles heal of cancer. Science 2002; 297: 63–64.

    Article  CAS  PubMed  Google Scholar 

  4. Sharma SV, Settleman J . Oncogene addiction: setting the stage for molecularly targeted cancer therapy. Genes Dev 2007; 21: 3214–3231.

    Article  CAS  PubMed  Google Scholar 

  5. Sharma SV, Bell DW, Settleman J, Haber DA . Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer 2007; 7: 169–181.

    Article  CAS  PubMed  Google Scholar 

  6. Neuzillet C, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S, Raymond E . MEK in cancer and cancer therapy. Pharmacol Ther 2014; 141: 160–171.

    Article  CAS  PubMed  Google Scholar 

  7. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature 2006; 439: 358–362.

    Article  CAS  PubMed  Google Scholar 

  8. Singh A, Greninger P, Rhodes D, Koopman L, Violette S, Bardeesy N et al. A gene expression signature associated with ‘K-Ras addiction’ reveals regulators of EMT and tumor cell survival. Cancer Cell 2009; 15: 489–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rebucci M, Michiels C . Molecular aspects of cancer cell resistance to chemotherapy. Biochem Pharmacol 2013; 85: 1219–1226.

    Article  CAS  PubMed  Google Scholar 

  10. Sos ML, Fischer S, Ullrich R, Peifer M, Heuckmann JM, Koker M et al. Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer. Proc Natl Acad Sci USA 2009; 106: 18351–18356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Klinger B, Sieber A, Fritsche-Guenther R, Witzel F, Berry L, Schumacher D et al. Network quantification of EGFR signaling unveils potential for targeted combination therapy. Mol Syst Biol 2013; 9: 673.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012; 483: 100–103.

    Article  CAS  PubMed  Google Scholar 

  13. Hatzivassiliou G, Haling JR, Chen H, Song K, Price S, Heald R et al. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature 2013; 501: 232–236.

    Article  CAS  PubMed  Google Scholar 

  14. Pratilas CA, Taylor BS, Ye Q, Viale A, Sander C, Solit DB et al. (V600E)BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway. Proc Natl Acad Sci USA 2009; 106: 4519–4524.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mirzoeva OK, Das D, Heiser LM, Bhattacharya S, Siwak D, Gendelman R et al. Basal subtype and MAPK/ERK kinase (MEK)-phosphoinositide 3-kinase feedback signaling determine susceptibility of breast cancer cells to MEK inhibition. Cancer Res 2009; 69: 565–572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med 2008; 14: 1351–1356.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bean GR, Ganesan YT, Dong Y, Takeda S, Liu H, Chan PM et al. PUMA and BIM are required for oncogene inactivation-induced apoptosis. Sci Signal 2013. 6–ra20.

  18. LoRusso P, Shapiro G, Pandya SS, Kwak EL, Jones C, Belvin M et al. A first-in-human phase Ib study to evaluate the MEK inhibitor GDC-0973, combined with the pan-PI3K inhibitor GDC-0941, in patients with advanced solid tumors. J Clin Oncol 2012. 30.

  19. Speranza G, Kinders RJ, Khin S, MK Weil, Do KT, Horneffer Y et al. Pharmacodynamic biomarker-driven trial of MK-2206, an AKT inhibitor, with AZD6244 (selumetinib), a MEK inhibitor, in patients with advanced colorectal carcinoma (CRC). J Clin Oncol 2012. 30.

  20. Komatsu N, Aoki K, Yamada M, Yukinaga H, Fujita Y, Kamioka Y et al. Development of an optimized backbone of FRET biosensors for kinases and GTPases. Mol Biol Cell 2011; 22: 4647–4656.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Friday BB, Yu C, Dy GK, Smith PD, Wang L, Thibodeau SN et al. BRAF V600E disrupts AZD6244-induced abrogation of negative feedback pathways between extracellular signal-regulated kinase and Raf proteins. Cancer Res 2008; 68: 6145–6153.

    Article  CAS  PubMed  Google Scholar 

  22. LoRusso PM, Krishnamurthi SS, Rinehart JJ, Nabell LM, Malburg L, Chapman PB et al. Phase I pharmacokinetic and pharmacodynamic study of the oral MAPK/ERK kinase inhibitor PD-0325901 in patients with advanced cancers. Clin Cancer Res 2010; 16: 1924–1937.

    Article  CAS  PubMed  Google Scholar 

  23. Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y et al. An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J Biol Chem 2009; 284: 8023–8032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. She QB, Halilovic E, Ye Q, Zhen W, Shirasawa S, Sasazuki T et al. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumors. Cancer Cell 2010; 18: 39–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rodriguez-Viciana P, Warne PH, Vanhaesebroeck B, Waterfield MD, Downward J . Activation of phosphoinositide 3-kinase by interaction with Ras and by point mutation. EMBO J 1996; 15: 2442–2451.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mendoza MC, Er EE, Blenis J . The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation. Trends Biochem Sci 2011; 36: 320–328.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Roux PP, Ballif BA, Anjum R, Gygi SP, Blenis J . Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase. Proc Natl Acad Sci USA 2004; 101: 13489–13494.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Carriere A, Romeo Y, Acosta-Jaquez HA, Moreau J, Bonneil E, Thibault P et al. ERK1/2 phosphorylate Raptor to promote Ras-dependent activation of mTOR complex 1 (mTORC1). J Biol Chem 2011; 286: 567–577.

    Article  CAS  PubMed  Google Scholar 

  29. Corcoran RB, Rothenberg SM, Hata AN, Faber AC, Piris A, Nazarian RM et al. TORC1 suppression predicts responsiveness to RAF and MEK inhibition in BRAF-mutant melanoma. Sci Transl Med 2013; 5 196ra198.

    Article  PubMed  Google Scholar 

  30. Yusa K, Rad R, Takeda J, Bradley A . Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon. Nat Methods 2009; 6: 363–369.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kawakami K, Noda T . Transposition of the Tol2 element, an Ac-like element from the Japanese medaka fish Oryzias latipes, in mouse embryonic stem cells. Genetics 2004; 166: 895–899.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Miyoshi H, Blomer U, Takahashi M, Gage FH, Verma IM . Development of a self-inactivating lentivirus vector. J Virol 1998; 72: 8150–8157.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Akagi T, Sasai K, Hanafusa H . Refractory nature of normal human diploid fibroblasts with respect to oncogene-mediated transformation. Proc Natl Acad Sci USA 2003; 100: 13567–13572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-Bromage H et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 2002; 110: 163–175.

    Article  CAS  PubMed  Google Scholar 

  35. Aoki K, Matsuda M . Visualization of small GTPase activity with fluorescence resonance energy transfer-based biosensors. Nat Protoc 2009; 4: 1623–1631.

    Article  CAS  PubMed  Google Scholar 

  36. Otsu N . A threshold selection method from gray-level histograms. IEEE Trans Syst Man Cybern 1979; 9: 62–66.

    Article  Google Scholar 

  37. Fujita Y, Komatsu N, Matsuda M, Aoki K . Fluorescence resonance energy transfer based quantitative analysis of feedforward and feedback loops in epidermal growth factor receptor signaling and the sensitivity to molecular targeting drugs. FEBS J 2014; 281: 3177–3192.

    Article  CAS  PubMed  Google Scholar 

  38. Corcoran RB, Dias-Santagata D, Bergethon K, Iafrate AJ, Settleman J, Engelman JA . BRAF gene amplification can promote acquired resistance to MEK inhibitors in cancer cells harboring the BRAF V600E mutation. Sci Signal 2010. 3–ra84.

  39. Aoki K, Kumagai Y, Sakurai A, Komatsu N, Fujita Y, Shionyu C et al. Stochastic ERK activation induced by noise and cell-to-cell propagation regulates cell density-dependent proliferation. Mol Cell 2013; 52: 529–540.

    Article  CAS  PubMed  Google Scholar 

  40. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B . Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 2008; 5: 621–628.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to the members of the Matsuda Laboratory for their helpful input. Y Inaoka, K Hirano, A Katsumata, N Nishimoto, N Nonaka and A Kawagishi are also to be thanked for their technical assistance. KA and MM were supported by the Research Program of Innovative Cell Biology by Innovative Technology (the ‘Cell Innovation’ Program) and Platform for Dynamic Approaches to Living System from the Ministry of Education, Culture, Sports and Science, Japan. KA was supported by a Grant-in-Aid for Young Scientists (B) (23701052) and a Grant-in-Aid for Scientific Research (B). NK and YF were supported by a Grant-in-Aid for JSPS Fellows.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to K Aoki.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Komatsu, N., Fujita, Y., Matsuda, M. et al. mTORC1 upregulation via ERK-dependent gene expression change confers intrinsic resistance to MEK inhibitors in oncogenic KRas-mutant cancer cells. Oncogene 34, 5607–5616 (2015). https://doi.org/10.1038/onc.2015.16

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2015.16

This article is cited by

Search

Quick links