Log in

Translational Therapeutics

The AKT inhibitor AZD5363 elicits synthetic lethality in ARID1A-deficient gastric cancer cells via induction of pyroptosis

  • Article
  • Published:
British Journal of Cancer Submit manuscript

Abstract

Background

Gastric cancer (GC) is a deadly disease with poor overall survival and limited therapeutic options. Genetic alterations such as mutations and/or deletions in chromatin remodeling gene AT-rich interactive domain 1 A (ARID1A) occur frequently in GC. Although ARID1A mutations/deletions are not a druggable target for conventional treatments, novel therapeutic strategies based on a synthetic lethal approach may be effective for the treatment of ARID1A-deficient cancers.

Methods

A kinase inhibitor library containing 551 compounds was screened in ARID1A isogenic GC cells for the ability to induce synthetic lethality effect. Selected hits’ activity was validated, and the mechanism of the most potent candidate drug, AKT inhibitor AD5363 (capivasertib), on induction of the synthetic lethality with ARID1A deficiency was investigated.

Results

After robust vulnerability screening of 551 diverse protein kinase inhibitors, we identified the AKT inhibitor AZD5363 as being the most potent lead compound in inhibiting viability of ARID1A−/− cells. A synthetic lethality between loss of ARID1A expression and AKT inhibition by AZD5363 was validated in both GC cell model system and xenograft model. Mechanistically, AZD5363 treatment induced pyroptotic cell death in ARID1A-deficient GC cells through activation of the Caspase-3/GSDME pathway. Furthermore, ARID1A occupied the AKT gene promoter and regulated its transcription negatively, thus the GC cells deficient in ARID1A showed increased expression and phosphorylation of AKT.

Conclusions

Our study demonstrates a novel synthetic lethality interaction and unique mechanism between ARID1A loss and AKT inhibition, which may provide a therapeutic and mechanistic rationale for targeted therapy on patients with ARID1A-defective GC who are most likely to be beneficial to AZD5363 treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Generation of ARID1A-knockout GC cells and screening of protein kinase inhibitor library for synthetic lethality.
Fig. 2: In vitro and in vivo synthetic lethality in ARID1A-KO GC cells by AKT inhibition.
Fig. 3: AZD5363 induces pyroptosis in ARID1A deficient cells.
Fig. 4: The pyroptosis induced by AZD5363 is mediated through caspase-3/GSDME pathway.
Fig. 5: ARID1A functions as AKT transcriptional repressor.
Fig. 6: A proposed model of the synthetic lethality between ARID1A loss and AKT inhibition.

Data availability

The authors agree to make the data in this paper publicly available on genuine request.

References

  1. Wu H, Wang W, Tong S, Wu C. Nucleostemin regulates proliferation and migration of gastric cancer and correlates with its malignancy. Int J Clin Exp Med. 2015;8:17634–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Ajani JA, Lee J, Sano T, Janjigian YY, Fan D, Song S. Gastric adenocarcinoma. Nat Rev Dis Primers. 2017;3:17036.

    Article  PubMed  Google Scholar 

  3. Harada K, Mizrak Kaya D, Shimodaira Y, Ajani JA. Global chemotherapy development for gastric cancer. Gastric Cancer. 2017;20:92–101.

    Article  CAS  PubMed  Google Scholar 

  4. Reisman D, Glaros S, Thompson EA. The SWI/SNF complex and cancer. Oncogene. 2009;28:1653–68.

    Article  CAS  PubMed  Google Scholar 

  5. Kadoch C, Hargreaves DC, Hodges C, Elias L, Ho L, Ranish J, et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet. 2013;45:592–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kadoch C, Crabtree GR. Reversible disruption of mSWI/SNF (BAF) complexes by the SS18-SSX oncogenic fusion in synovial sarcoma. Cell. 2013;153:71–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wilson BG, Wang X, Shen X, McKenna ES, Lemieux ME, Cho YJ, et al. Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell. 2010;18:316–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. McBride MJ, Pulice JL, Beird HC, Ingram DR, D’Avino AR, Shern JF, et al. The SS18-SSX Fusion Oncoprotein Hijacks BAF Complex Targeting and Function to Drive Synovial Sarcoma. Cancer Cell. 2018;33:1128–41.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zang ZJ, Cutcutache I, Poon SL, Zhang SL, McPherson JR, Tao J, et al. Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet. 2012;44:570–4.

    Article  CAS  PubMed  Google Scholar 

  10. Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.

    Article  CAS  PubMed  Google Scholar 

  11. Bass AJ, Thorsson V, Shmulevich I, Reynolds SM, Miller M, Bernard B, et al. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.

  12. Li J, Wang W, Zhang Y, Cieślik M, Guo J, Tan M, et al. Epigenetic driver mutations in ARID1A shape cancer immune phenotype and immunotherapy. J Clin Invest. 2020;130:2712–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lord CJ, Ashworth A. PARP inhibitors: Synthetic lethality in the clinic. Science. 2017;355:1152–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Pilié PG, Tang C, Mills GB, Yap TA. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol. 2019;16:81–104.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med. 2018;24:556–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Li T, Shi L, Liu W, Hu X, Hui Y, Di M, et al. Aloe-Emodin Induces Mitochondrial Dysfunction and Pyroptosis by Activation of the Caspase-9/3/Gasdermin E Axis in HeLa Cells. Front Pharmacol. 2022;13:854526.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Andrikopoulou A, Chatzinikolaou S, Panourgias E, Kaparelou M, Liontos M, Dimopoulos MA, et al. The emerging role of capivasertib in breast cancer. Breast. 2022;63:157–67.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Jiang M, Qi L, Li L, Li Y. The caspase-3/GSDME signal pathway as a switch between apoptosis and pyroptosis in cancer. Cell Death Discov. 2020;6:112.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Tan Y, Chen Q, Li X, Zeng Z, **ong W, Li G, et al. Pyroptosis: a new paradigm of cell death for fighting against cancer. J Exp Clin Cancer Res. 2021;40:153.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang Y, Gao W, Shi X, Ding J, Liu W, He H, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547:99–103.

    Article  CAS  PubMed  Google Scholar 

  21. Rogers C, Fernandes-Alnemri T, Mayes L, Alnemri D, Cingolani G, Alnemri ES. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat Commun. 2017;8:14128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dong S, Shi Y, Dong X, **ao X, Qi J, Ren L, et al. Gasdermin E is required for induction of pyroptosis and severe disease during enterovirus 71 infection. J Biol Chem. 2022;298:101850.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lee D, Yu EJ, Ham IH, Hur H, Kim YS. AKT inhibition is an effective treatment strategy in ARID1A-deficient gastric cancer cells. Onco Targets Ther. 2017;10:4153–9.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Samartzis EP, Noske A, Dedes KJ, Fink D, Imesch P. ARID1A mutations and PI3K/AKT pathway alterations in endometriosis and endometriosis-associated ovarian carcinomas. Int J Mol Sci. 2013;14:18824–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wilson BG, Roberts CW. SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer. 2011;11:481–92.

    Article  CAS  PubMed  Google Scholar 

  26. Oike T, Ogiwara H, Tominaga Y, Ito K, Ando O, Tsuta K, et al. A synthetic lethality-based strategy to treat cancers harboring a genetic deficiency in the chromatin remodeling factor BRG1. Cancer Res. 2013;73:5508–18.

    Article  CAS  PubMed  Google Scholar 

  27. Helming KC, Wang X, Wilson BG, Vazquez F, Haswell JR, Manchester HE, et al. ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nat Med. 2014;20:251–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;4:988–1004.

    Article  CAS  PubMed  Google Scholar 

  29. Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 2006;441:424–30.

    Article  CAS  PubMed  Google Scholar 

  30. Li L, Wang S, Zhou W. Balance Cell Apoptosis and Pyroptosis of Caspase-3-Activating Chemotherapy for Better Antitumor Therapy. Cancers (Basel). 2022;15:26.

    Article  PubMed  Google Scholar 

  31. Kim YB, Ham IH, Hur H, Lee D. Various ARID1A expression patterns and their clinical significance in gastric cancers. Hum Pathol. 2016;49:61–70.

    Article  CAS  PubMed  Google Scholar 

  32. Chandler RL, Brennan J, Schisler JC, Serber D, Patterson C, Magnuson T. ARID1a-DNA interactions are required for promoter occupancy by SWI/SNF. Mol Cell Biol. 2013;33:265–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge the central animal facility at Fujian Medical University for the help with animal experiments. We sincerely thank the technical support from Jun** Lin and Shu** Zheng (Public Technology Service Center, Fujian Medical University) for assistance with flow cytometry and confocal microscopy experiments.

Funding

This work was supported in part by a grant from the National Natural Science Foundation of China (81672967), the Startup Fund for High-Level Talents of Fujian Medical University (XRCZX2017021), and the Startup Fund for Scientific Research of Fujian Medical University (No. 2019QH2003).

Author information

Authors and Affiliations

Authors

Contributions

MHF and YFL designed and performed experiments, analyzed data and wrote the manuscript. CRX, KQS, ZGG, YTH, HBL, YCW, and YCS performed the experiments and analyzed data. SBH and XTC contributed to data interpretation and editing the paper. XL and XJL designed experiments, supervised the study, wrote and edit the manuscript.

Corresponding authors

Correspondence to Xu Lin or **njian Lin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

This article does not contain any studies with human participants. All procedures were carried out according to the Animal Care and Use Committee protocol approved for this study by Fujian Medical University (FMU-IACUC 15–066) and performed in accordance with the institution guidelines.

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

Fang, M., Lin, Y., Xue, C. et al. The AKT inhibitor AZD5363 elicits synthetic lethality in ARID1A-deficient gastric cancer cells via induction of pyroptosis. Br J Cancer (2024). https://doi.org/10.1038/s41416-024-02778-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41416-024-02778-5

  • Springer Nature Limited

Navigation