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:

Induction of SREBP1 degradation coupled with suppression of SREBP1-mediated lipogenesis impacts the response of EGFR mutant NSCLC cells to osimertinib

Abstract

Emergence of acquired resistance to osimertinib (AZD9291), the first-approved third-generation EGFR inhibitor that selectively and irreversibly inhibits the activating EGFR mutations and the resistant T790M mutation, is a giant and urgent clinical challenge. Fully understanding the biology underlying the response of EGFR mutant non-small cell lung cancer (NSCLC) to osimertinib is the foundation for development of mechanism-driven strategies to overcome acquired resistance to osimertinib or other third-generation EGFR inhibitors. This study focused on tackling this important issue by elucidating the critical role of sterol regulatory element-binding protein 1 (SREBP1) degradation in conferring the response of EGFR mutant NSCLC cells to osimertinib and by validating the strategy via directly targeting SREBP1 for overcoming osimertinib acquired resistance. Osimertinib facilitated degradation of the mature form of SREBP1 (mSREBP1) in a GSK3/FBXW7-dependent manner and reduced protein levels of its regulated genes in EGFR-mutant NSCLC cells/tumors accompanied with suppression of lipogenesis. Once resistant, EGFR-mutant NSCLC cell lines possessed elevated levels of mSREBP1, which were resistant to osimertinib modulation. Both genetic and pharmacological inhibition of SREBP1 sensitized osimertinib-resistant cells and tumors to osimertinib primarily through enhancing Bim-dependent induction of apoptosis, whereas enforced expression of ectopic SREBP1 in sensitive EGFR-mutant NSCLC cells compromised osimertinib’s cell-killing effects. Collectively, we have demonstrated a novel connection between osimertinib and SREBP1 degradation and its impact on the response of EGFR mutant NSCLC cells to osimertinib and suggested an effective strategy for overcoming acquired resistance to osimertinib, and possibly other EGFR inhibitors, via targeting SREBP1.

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: Osimertinib inhibits mTORC2 signaling and decreases mSREBP1 levels in EGFRm NSCLC cells accompanied with reduction of ACC and FASN.
Fig. 2: Osimertinib effectively suppresses SREBP1-regulated lipid metabolism in EGFRm NSCLC cells.
Fig. 3: Osimertinib reduces mSREBP1 levels through facilitating GSK3/FBXW7-mediated mSREBP1 degradation.
Fig. 4: EGFRm NSCLC cells and tissues with acquired resistance to osimertinib or other EGFR-TKIs possess elevated levels of mSREBP1 and/or FASN, which are resistant to osimertinib modulation, and enforced expression of ectopic in sensitive EGFRm NSCLC cells confers resistance to osimertinib.
Fig. 5: Genetic knockdown of SREBP1 reverses osimertinib resistance in vitro and in vivo.
Fig. 6: Chemical inhibition of SREBP1 combined with osimertinib synergistically decreases survival with enhanced apoptosis of osimertinib-resistant NSCLC cells and overcomes osimertinib resistance in vivo.
Fig. 7: Osimertinib combined with SREBP1 inhibition enhances Bim and Mcl-1 levels and augments Bim-dependent apoptosis in osimertinib-resistant cells.

Similar content being viewed by others

References

  1. Malvezzi M, Carioli G, Bertuccio P, Boffetta P, Levi F, La Vecchia C, et al. European cancer mortality predictions for the year 2017, with focus on lung cancer. Ann Oncol. 2017;28:1117–23.

    Article  CAS  Google Scholar 

  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.

    Article  Google Scholar 

  3. Recondo G, Facchinetti F, Olaussen KA, Besse B, Friboulet L. Making the first move in EGFR-driven or ALK-driven NSCLC: first-generation or next-generation TKI? Nat Rev Clin Oncol. 2018;15:694–708.

    Article  CAS  Google Scholar 

  4. Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N. Engl J Med. 2018;378:113–25.

    Article  CAS  Google Scholar 

  5. Röhrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 2016;16:732–49.

    Article  Google Scholar 

  6. Merino Salvador M, Gómez de Cedrón M, Moreno Rubio J, Falagán Martínez S, Sánchez Martínez R, Casado E, et al. Lipid metabolism and lung cancer. Crit Rev Oncol/Hematol. 2017;112:31–40.

    Article  Google Scholar 

  7. Guo D, Reinitz F, Youssef M, Hong C, Nathanson D, Akhavan D, et al. An LXR agonist promotes glioblastoma cell death through inhibition of an EGFR/AKT/SREBP-1/LDLR-dependent pathway. Cancer Discov. 2011;1:442–56.

    Article  CAS  Google Scholar 

  8. Yue S, Li J, Lee SY, Lee HJ, Shao T, Song B, et al. Cholesteryl ester accumulation induced by PTEN loss and PI3K/AKT activation underlies human prostate cancer aggressiveness. Cell Metab. 2014;19:393–406.

    Article  CAS  Google Scholar 

  9. Guo D, Bell EH, Mischel P, Chakravarti A. Targeting SREBP-1-driven lipid metabolism to treat cancer. Curr Pharm Des. 2014;20:2619–26.

    Article  CAS  Google Scholar 

  10. Cheng X, Li J, Guo D. SCAP/SREBPs are Central Players in Lipid Metabolism and Novel Metabolic Targets in Cancer Therapy. Curr Top Med Chem. 2018;18:484–93.

    Article  CAS  Google Scholar 

  11. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Investig. 2002;109:1125–31.

    Article  CAS  Google Scholar 

  12. Guo D, Prins RM, Dang J, Kuga D, Iwanami A, Soto H, et al. EGFR signaling through an Akt-SREBP-1-dependent, rapamycin-resistant pathway sensitizes glioblastomas to antilipogenic therapy. Sci Signal. 2009;2:ra82.

    Article  Google Scholar 

  13. Shi P, Oh YT, Deng L, Zhang G, Qian G, Zhang S, et al. Overcoming acquired resistance to AZD9291, a third-generation EGFR inhibitor, through modulation of MEK/ERK-dependent Bim and Mcl-1 degradation. Clin Cancer Res. 2017;23:6567–79.

    Article  CAS  Google Scholar 

  14. Wee P, Wang Z. Epidermal growth factor receptor cell proliferation signaling pathways. Cancers. 2017;9:52.

    Article  Google Scholar 

  15. Oh WJ, Jacinto E. mTOR complex 2 signaling and functions. Cell Cycle. 2011;10:2305–16.

    Article  CAS  Google Scholar 

  16. Li S, Oh YT, Yue P, Khuri FR, Sun SY. Inhibition of mTOR complex 2 induces GSK3/FBXW7-dependent degradation of sterol regulatory element-binding protein 1 (SREBP1) and suppresses lipogenesis in cancer cells. Oncogene. 2016;35:642–50.

    Article  CAS  Google Scholar 

  17. Hagiwara A, Cornu M, Cybulski N, Polak P, Betz C, Trapani F, et al. Hepatic mTORC2 activates glycolysis and lipogenesis through Akt, glucokinase, and SREBP1c. Cell Metab. 2012;15:725–38.

    Article  CAS  Google Scholar 

  18. Ali A, Levantini E, Teo JT, Goggi J, Clohessy JG, Wu CS, et al. Fatty acid synthase mediates EGFR palmitoylation in EGFR mutated non-small cell lung cancer. EMBO Mol Med. 2018;10:e8313.

    Article  Google Scholar 

  19. Polonio-Alcala E, Palomeras S, Torres-Oteros D, Relat J, Planas M, Feliu L, et al. Fatty acid synthase inhibitor G28 shows anticancer activity in EGFR tyrosine kinase inhibitor resistant lung adenocarcinoma models. Cancers. 2020;12:1283.

    Article  CAS  Google Scholar 

  20. Zhou J, Qu G, Zhang G, Wu Z, Liu J, Yang D, et al. Glycerol kinase 5 confers gefitinib resistance through SREBP1/SCD1 signaling pathway. J Exp Clin Cancer Res. 2019;38:96.

    Article  Google Scholar 

  21. Li J, Yan H, Zhao L, Jia W, Yang H, Liu L, et al. Inhibition of SREBP increases gefitinib sensitivity in non-small cell lung cancer cells. Oncotarget. 2016;7:52392–403.

    Article  Google Scholar 

  22. Li Y, Zang H, Qian G, Owonikoko TK, Ramalingam SR. ERK inhibition effectively overcomes acquired resistance of epidermal growth factor receptor-mutant non-small cell lung cancer cells to osimertinib. Cancer. 2020;126:1339–50.

    Article  CAS  Google Scholar 

  23. Zhao W, Yu D, Chen Z, Yao W, Yang J, Ramalingam SS, et al. Inhibition of MEK5/ERK5 signaling overcomes acquired resistance to the third generation EGFR inhibitor, osimertinib, via enhancing Bim-dependent apoptosis. Cancer Lett. 2021;519:141–9.

    Article  CAS  Google Scholar 

  24. Zang H, Qian G, Arbiser J, Owonikoko TK, Ramalingam SS, Fan S, et al. Overcoming acquired resistance of EGFR-mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib, with the natural product honokiol. Mol Oncol. 2020;14:882–95.

    Article  CAS  Google Scholar 

  25. Yu D, Li Y, Sun KD, Gu J, Chen Z, Owonikoko TK, et al. The novel MET inhibitor, HQP8361, possesses single agent activity and enhances therapeutic efficacy of AZD9291 (osimertinib) against AZD9291-resistant NSCLC cells with activated MET. Am J Cancer Res. 2020;10:3316–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Sun SY, Yue P, Dawson MI, Shroot B, Michel S, Lamph WW, et al. Differential effects of synthetic nuclear retinoid receptor-selective retinoids on the growth of human non-small cell lung carcinoma cells. Cancer Res. 1997;57:4931–9.

    CAS  PubMed  Google Scholar 

  27. Yao W, Yue P, Zhang G, Owonikoko TK, Khuri FR, Sun SY. Enhancing therapeutic efficacy of the MEK inhibitor, MEK162, by blocking autophagy or inhibiting PI3K/Akt signaling in human lung cancer cells. Cancer Lett. 2015;364:70–78.

    Article  CAS  Google Scholar 

  28. Geng F, Cheng X, Wu X, Yoo JY, Cheng C, Guo JY, et al. Inhibition of SOAT1 suppresses glioblastoma growth via blocking SREBP-1-mediated lipogenesis. Clin Cancer Res. 2016;22:5337–48.

    Article  CAS  Google Scholar 

  29. Takada M, Zhang W, Suzuki A, Kuroda TS, Yu Z, Inuzuka H, et al. FBW7 loss promotes chromosomal instability and tumorigenesis via cyclin E1/CDK2-mediated phosphorylation of CENP-A. Cancer Res. 2017;77:4881–93.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are thankful to Dr. Dongsheng Wang and Mr. Guojing Zhang in our department for helping us with animal experiments, to Dr. Teng Wang and Dr. Dong Hua (Affiliated Hospital of Jiangnan University) for providing us with clinical samples, to Dr. Peng Zhang (Longgang E.N.T. Hospital & Shenzhen Key Laboratory of E.N.T.) for staining the patients’ samples, and to Drs. Deliang Guo and Wenyi Wei for providing some shRNAs. We are also grateful to Dr. Anthea Hammond in our department for editing the manuscript. This work was supported by the NIH/NCI R01 CA223220 (to S-YS), R01 CA245386 (to S-YS), UG1 CA233259 (to SSR) and lung cancer SPORE P50 CA217691 DRP award (to S-YS) and Emory University Winship Cancer Institute lung cancer pilot funds (to S-YS). DY is a visiting medical student participating in the Xiangya-Emory Visiting Medical Student Program. SSR, TKO, and S-YS are Georgia Research Alliance Distinguished Cancer Scientists.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: S-YS, ZC, TKO, and SSR. Investigation: ZC and DY. Methodology: ZC and S-YS. Writing/Original draft: ZC and S-YS. Funding acquisition: S-YS and SSR. Supervision: TKO, SSR, and S-YS.

Corresponding author

Correspondence to Shi-Yong Sun.

Ethics declarations

Competing interests

SSR is on consulting/advisory board for AstraZeneca, Amgen, BMS, Merck, Roche, Tesaro and Amgen. TKO is on consulting/advisory board for Novartis, Celgene, Lilly, Sandoz, Abbvie, Eisai, Takeda, Bristol-Myers Squibb, MedImmune, Amgen, AstraZeneca and Boehringer Ingelheim. Other authors declare that they have no conflict of interest.

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Z., Yu, D., Owonikoko, T.K. et al. Induction of SREBP1 degradation coupled with suppression of SREBP1-mediated lipogenesis impacts the response of EGFR mutant NSCLC cells to osimertinib. Oncogene 40, 6653–6665 (2021). https://doi.org/10.1038/s41388-021-02057-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-021-02057-0

This article is cited by

Search

Quick links