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.

  • Article
  • Published:

Translational Therapeutics

PAK and PI3K pathway activation confers resistance to KRASG12C inhibitor sotorasib

Abstract

Background

KRAS is a frequently mutated oncogene in human cancer. Clinical studies on the covalent inhibitors of the KRASG12C mutant have reported promising results. However, primary and acquired resistance may limit their clinical use.

Methods

Sotorasib-resistant cell lines were established. We explored the signalling pathways activated in these resistant cell lines and their roles in sotorasib resistance.

Results

The resistant cells exhibited increased cell–matrix adhesion with increased levels of stress fibres and focal adherens. p21-activated kinases (PAKs) were activated in resistant cells, which phosphorylate MEK at serine 298 of MEK and serine 338 of c-Raf to activate the mitogen-activated protein kinase pathway. The PAK inhibitors FRAX597 and FRAX486 in synergy with sotorasib reduced the viability of KRASG12C mutant cancer cells. Furthermore, the PI3K/AKT pathway was constitutively active in sotorasib-resistant cells. The overexpression of constitutively activated PI3K or the knockdown of PTEN resulted in resistance to sotorasib. PI3K inhibitor alpelisib was synergistic with sotorasib in compromising the viability of KRASG12C mutant cancer cells. Moreover, PI3K and PAK pathways formed a mutual positive regulatory loop that mediated sotorasib resistance.

Conclusions

Our results indicate that the cell–matrix interaction-dependent activation of PAK mediates resistance to sotorasib through the activation of MAPK and PI3K pathways.

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

Fig. 1: Establishment of sotorasib-resistant cancer cell lines.
Fig. 2: Sotorasib-resistant cancer cells exhibit enhanced cell–matrix adhesion.
Fig. 3: Activation of the Src/PAK/MEK axis in sotorasib-resistant cells.
Fig. 4: PAK inhibitors and sotorasib synergistically kill cancer cells.
Fig. 5: Activation of the PI3K pathway confers resistance to sotorasib.
Fig. 6: Mutual positive regulatory loop between the PI3K and PAK pathways regulates cellular sensitivity to sotorasib and the antitumour effects of the combination of sotorasib with FRAX597 or alpelisib in vivo.

Similar content being viewed by others

Data availability

The mRNA sequencing data were deposited to the National Center for Biotechnology Information Genome Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE178479. Other data sets generated and/or analysed during this study are available from the corresponding author on reasonable request.

References

  1. Rodenhuis S. Ras and human tumors. Semin Cancer Biol. 1992;3:241–7.

    CAS  Google Scholar 

  2. Andreyev HJ, Norman AR, Cunningham D, Oates J, Dix BR, Iacopetta BJ, et al. Kirsten ras mutations in patients with colorectal cancer: the ‘RASCAL II’ study. Br J Cancer. 2001;85:692–6.

    Article  CAS  Google Scholar 

  3. Milburn MV, Tong L, deVos AM, Brünger A, Yamaizumi Z, Nishimura S, et al. Molecular switch for signal transduction: structural differences between active and inactive forms of protooncogenic ras proteins. Science. 1990;247:939–45.

    Article  CAS  Google Scholar 

  4. Nollmann FI, Ruess DA. Targeting mutant KRAS in pancreatic cancer: Futile or promising? Biomedicines. 2020;8:E281.

    Article  Google Scholar 

  5. Janes MR, Zhang J, Li LS, Hansen R, Peters U, Guo X, et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell. 2018;172:578–89.

    Article  CAS  Google Scholar 

  6. Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The clinical KRASG12C inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575:217–23.

    Article  CAS  Google Scholar 

  7. Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, et al. The KRASG12C inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients. Cancer Discov. 2020;10:54–71.

    Article  CAS  Google Scholar 

  8. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-RasG12C inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–51.

    Article  CAS  Google Scholar 

  9. Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRAS G12C inhibition with Sotorasib in advanced solid tumors. N Engl J Med. 2020;383:1207–17.

    Article  CAS  Google Scholar 

  10. Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, et al. Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med. 2021;384:2371–81.

    Article  CAS  Google Scholar 

  11. Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, et al. Acquired resistance to KRAS(G12C) inhibition in cancer. N Engl J Med. 2021;384:2382–93.

    Article  CAS  Google Scholar 

  12. Zhao Y, Murciano-Goroff YR, Xue JY, Ang A, Lucas J, Mai TT, et al. Diverse alterations associated with resistance to KRAS(G12C) inhibition. Nature. 2021;599:679–83.

    Article  CAS  Google Scholar 

  13. Ryan MB, Fece de la Cruz F, Phat S, Myers DT, Wong E, Shahzade HA, et al. Vertical pathway inhibition overcomes adaptive feedback resistance to KRASG12C Inhibition. Clin Cancer Res. 2020;26:1633–43.

    Article  CAS  Google Scholar 

  14. Adachi Y, Ito K, Hayashi Y, Kimura R, Tan TZ, Yamaguchi R, et al. Epithelial to mesenchymal transition is a cause of both intrinsic and acquired resistance to KRAS G12C inhibitor in KRAS G12C mutant non-small cell lung cancer. Clin Cancer Res. 2020;26:5962–73.

    Article  CAS  Google Scholar 

  15. Xue JY, Zhao Y, Aronowitz J, Mai TT, Vides A, Qeriqi B, et al. Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition. Nature 2020;577:421–5.

    Article  CAS  Google Scholar 

  16. Amodio V, Yaeger R, Arcella P, Cancelliere C, Lamba S, Lorenzato A, et al. EGFR blockade reverts resistance to KRAS(G12C) inhibition in colorectal cancer. Cancer Discov. 2020;10:1129–39.

    Article  CAS  Google Scholar 

  17. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.

    Article  Google Scholar 

  18. Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul. 1984;22:27–55.

    Article  CAS  Google Scholar 

  19. Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, Lopez M, et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci USA. 2010;107:8788–93.

    Article  CAS  Google Scholar 

  20. Beeser A, Jaffer ZM, Hofmann C, Chernoff J. Role of group A p21-activated kinase in activation of extracellular-regulated kinase by growth factors. J Biol Chem. 2005;280:36609–15.

    Article  CAS  Google Scholar 

  21. Chaudhary A, King WG, Mattaliano MD, Frost JA, Diaz B, Morrison DK, et al. Phosphatidylinositol 3-kinase regulates Raf1 through Pak phosphorylation of serine 338. Curr Biol. 2000;10:551–4.

    Article  CAS  Google Scholar 

  22. Manser E, Leung T, Salihuddin H, Zhao ZS, Lim L. A brain serine/threonine protein kinase activated by Cdc42 and Rac1. Nature 1994;367:40–6.

    Article  CAS  Google Scholar 

  23. Niit M, Hoskin V, Carefoot E, Geletu M, Arulanandam R, Elliott B, et al. Cell-cell and cell-matrix adhesion in survival and metastasis: Stat3 versus Akt. Biomol Concepts. 2015;6:383–99.

    Article  CAS  Google Scholar 

  24. Brown MC, Cary LA, Jamieson JS, Cooper JA, Turner CE. Src and FAK kinases cooperate to phosphorylate paxillin kinase linker, stimulate its focal adhesion localization, and regulate cell spreading and protrusiveness. Mol Biol Cell. 2005;16:4316–28.

    Article  CAS  Google Scholar 

  25. Slack-Davis JK, Eblen ST, Zecevic M, Boerner SA, Tarcsafalvi A, Diaz HB, et al. PAK1 phosphorylation of MEK1 regulates fibronectin-stimulated MAPK activation. J Cell Biol. 2003;162:281–91.

    Article  CAS  Google Scholar 

  26. Harris KF, Shoji I, Cooper EM, Kumar S, Oda H, Howley PM. Ubiquitin-mediated degradation of active Src tyrosine kinase. Proc Natl Acad Sci USA. 1999;96:13738–43.

    Article  CAS  Google Scholar 

  27. Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell. 2007;129:1261–74.

    Article  CAS  Google Scholar 

  28. André F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N. Engl J Med. 2019;380:1929–40.

    Article  Google Scholar 

  29. Higuchi M, Onishi K, Kikuchi C, Gotoh Y. Scaffolding function of PAK in the PDK1-Akt pathway. Nat Cell Biol. 2008;10:1356–64.

    Article  CAS  Google Scholar 

  30. Zhou GL, Zhuo Y, King CC, Fryer BH, Bokoch GM, Field J. Akt phosphorylation of serine 21 on Pak1 modulates Nck binding and cell migration. Mol Cell Biol. 2003;23:8058–69.

    Article  CAS  Google Scholar 

  31. Rane CK, Minden A. P21 activated kinase signaling in cancer. Semin Cancer Biol. 2019;54:40–9.

    Article  CAS  Google Scholar 

  32. Sinha S, Yang W. Cellular signaling for activation of Rho GTPase Cdc42. Cell Signal. 2008;20:1927–34.

    Article  CAS  Google Scholar 

  33. Zhang B, Zhang Y, Zhang J, Liu P, Jiao B, Wang Z, et al. Focal adhesion kinase (FAK) inhibition synergizes with KRAS G12C inhibitors in treating cancer through the regulation of the FAK-YAP signaling. Adv Sci. 2021;8:e2100250.

  34. Chung EY, Mai Y, Shah UA, Wei Y, Ishida E, Kataoka K, et al. PAK kinase inhibition has therapeutic activity in novel preclinical models of adult T-cell leukemia/lymphoma. Clin Cancer Res. 2019;25:3589–601.

    Article  CAS  Google Scholar 

  35. Semenova G, Chernoff J. Targeting PAK1. Biochem Soc Trans. 2017;45:79–88.

    Article  CAS  Google Scholar 

  36. Lu H, Liu S, Zhang G, Wu B, Zhu Y, Frederick DT, et al. PAK signalling drives acquired drug resistance to MAPK inhibitors in BRAF-mutant melanomas. Nature. 2017;550:133–6.

    Article  CAS  Google Scholar 

  37. Brazil DP, Yang ZZ, Hemmings BA. Advances in protein kinase B signalling: AKTion on multiple fronts. Trends Biochem Sci. 2004;29:233–42.

    Article  CAS  Google Scholar 

  38. Downward J. PI 3-kinase, Akt and cell survival. Semin Cell Dev Biol. 2004;15:177–82.

    Article  CAS  Google Scholar 

  39. Jabbarzadeh Kaboli P, Salimian F, Aghapour S, Xiang S, Zhao Q, Li M, et al. Akt-targeted therapy as a promising strategy to overcome drug resistance in breast cancer - a comprehensive review from chemotherapy to immunotherapy. Pharm Res. 2020;156:104806.

    Article  CAS  Google Scholar 

  40. Mayer IA, Arteaga CL. The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med. 2016;67:11–28.

    Article  CAS  Google Scholar 

  41. Misale S, Fatherree JP, Cortez E, Egan RK, McClanaghan J, Stein GT, et al. KRAS G12C NSCLC models are sensitive to direct targeting of KRAS in combination with PI3K inhibition. Clin Cancer Res. 2019;25:796–807.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge technical support from the Second and Eighth Core Labs of National Taiwan University Hospital, the Translational Core Facility of Taipei Medical University, and RNA Technology Platform and Gene Manipulation Core, Academia Sinica.

Funding

This work was supported by the Ministry of Science and Technology, Taiwan [grant numbers 109-2314-B-002-084 and 110-2314-B-002-173].

Author information

Authors and Affiliations

Authors

Contributions

C-YY and Y-MJ conceptualised the study and designed the experiments and provided financial support. C-HC, L-WC, and T-YL performed the experiments. Y-RL and T-HH collected the bioinformatics data. C-YY, L-WC, and Y-MJ interpreted the data and drafted the manuscript. All authors discussed the results; reviewed and revised the manuscript; and approved the final version of the manuscript.

Corresponding authors

Correspondence to Ching-Yao Yang or Yung-Ming Jeng.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chan, CH., Chiou, LW., Lee, TY. et al. PAK and PI3K pathway activation confers resistance to KRASG12C inhibitor sotorasib. Br J Cancer 128, 148–159 (2023). https://doi.org/10.1038/s41416-022-02032-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-022-02032-w

This article is cited by

Search

Quick links