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

Characterisation of a novel KRAS G12C inhibitor ASP2453 that shows potent anti-tumour activity in KRAS G12C-mutated preclinical models

Abstract

Background

KRAS is one of the most frequently mutated oncogenes in various cancers, and several novel KRAS G12C direct inhibitors are now in clinical trials. Here, we characterised the anti-tumour efficacy of ASP2453, a novel KRAS G12C inhibitor, in preclinical models of KRAS G12C-mutated cancer.

Methods

We evaluated the in vitro and in vivo activity of ASP2453, alone or in combination with targeted agents and immune checkpoint inhibitors, in KRAS G12C-mutated cancer cells and xenograft models. We also assessed pharmacological differences between ASP2453 and AMG 510, another KRAS G12C inhibitor, using an SPR assay, washout experiments and an AMG 510-resistant xenograft model.

Results

ASP2453 potently and selectively inhibited KRAS G12C-mediated growth, KRAS activation and downstream signalling in vitro and in vivo, and improved the anti-tumour effects of targeted agents and immune checkpoint inhibitors. Further, ASP2453 had more rapid binding kinetics to KRAS G12C protein and showed more potent inhibitory effects on KRAS activation and cell proliferation after washout than AMG 510. ASP2453 also induced tumour regression in an AMG 510-resistant xenograft model.

Conclusions

ASP2453 is a potential therapeutic agent for KRAS G12C-mutated cancer. ASP2453 showed efficacy in AMG 510-resistant tumours, even among compounds with the same mode of action.

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: ASP2453 is a potent, selective and covalent KRAS G12C inhibitor.
Fig. 2: ASP2453 selectively inhibits KRAS signalling and proliferation of KRAS G12C-mutated in cancer cells in vitro.
Fig. 3: Oral administration of ASP2453 induces anti-tumour activity in an NCI-H1373 xenograft model.
Fig. 4: ASP2453 shows potent anti-tumour activity in multiple human cancer xenograft models.
Fig. 5: Combination ASP2453 with erlotinib or anti-PD-1 antibody inhibits cell proliferation in vitro and tumour growth in vivo.
Fig. 6: Comparison of ASP2453 and AMG 510 in vitro and in vivo.

Similar content being viewed by others

Data availability

Researchers may request access to anonymized participant-level data, trial-level data and protocols from Astellas sponsored clinical trials at www.clinicalstudydatarequest.com. For the Astellas criteria on data sharing see https://clinicalstudydatarequest.com/Study-Sponsors/Study-Sponsors-Astellas.aspx.

References

  1. Wennerberg K, Rossman KL, Der CJ. The Ras superfamily at a glance. J Cell Sci. 2005;118:843–6.

    Article  CAS  Google Scholar 

  2. Ryan MB, Corcoran RB. Therapeutic strategies to target RAS-mutant cancers. Nat Rev Clin Oncol. 2018;15:709–20.

    Article  CAS  Google Scholar 

  3. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer. 2011;11:761–74.

    Article  CAS  Google Scholar 

  4. Cox AD, Fesik SW, Kimmelman AC, Luo J, Der CJ. Drugging the undruggable RAS: Mission possible? Nat Rev Drug Discov. 2014;13:828–51.

    Article  CAS  Google Scholar 

  5. Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol Cancer Res. 2015;13:1325–35.

    Article  CAS  Google Scholar 

  6. Guerrero S, Casanova I, Farré L, Mazo A, Capellà G, Mangues R. K-ras codon 12 mutation induces higher level of resistance to apoptosis and predisposition to anchorage-independent growth than codon 13 mutation or proto-oncogene overexpression. Cancer Res. 2000;60:6750–6.

    CAS  PubMed  Google Scholar 

  7. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–50.

    Article  Google Scholar 

  8. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–386.

    Article  CAS  Google Scholar 

  9. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7–30.

    Article  Google Scholar 

  10. Ahrendt SA, Decker PA, Alawi EA, Zhu Y-r, Sanchez-Cespedes M, Yang SC, et al. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung. Cancer. 2001;92:1525–30.

    Article  CAS  Google Scholar 

  11. Dogan S, Shen R, Ang DC, Johnson ML, D’Angelo SP, Paik PK, et al. Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung adenocarcinomas: higher susceptibility of women to smoking-related KRAS-mutant cancers. Clin Cancer Res. 2012;18:6169–77.

    Article  CAS  Google Scholar 

  12. Nadal E, Chen G, Prensner JR, Shiratsuchi H, Sam C, Zhao L, et al. KRAS-G12C mutation is associated with poor outcome in surgically resected lung adenocarcinoma. J Thorac Oncol. 2014;9:1513–22.

    Article  CAS  Google Scholar 

  13. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–51.

    Article  CAS  Google Scholar 

  14. Lim SM, Westover KD, Ficarro SB, Harrison RA, Choi HG, Pacold ME, et al. Therapeutic targeting of oncogenic K-Ras by a covalent catalytic site inhibitor. Angew Chem Int Ed Engl. 2014;53:199–204.

    Article  CAS  Google Scholar 

  15. Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575:217–23.

    Article  CAS  Google Scholar 

  16. Lanman BA, Allen JR, Allen JG, Amegadzie AK, Ashton KS, Booker SK, et al. Discovery of a covalent inhibitor of KRAS(G12C) (AMG 510) for the treatment of solid tumors. J Med Chem. 2020;63:52–65.

    Article  CAS  Google Scholar 

  17. Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, et al. The KRAS(G12C) inhibitor MRTX849 provides Insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients. Cancer Disco. 2020;10:54–71.

    Article  CAS  Google Scholar 

  18. 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 

  19. Jänne, PA, Rybkin, II, Spira, AI, Riely, GJ, Papadopoulos, KP, Sabari, JK, et al. KRYSTAL-1: activity and safety of adagrasib (MRTX849) in advanced/metastatic non–small-cell lung cancer (NSCLC) harboring KRAS G12C mutation. Presented at the 2020 AACR-NCI-EORTC meeting (2020).

  20. Johnson, ML, Ou, SHI, Barve, M, Rybkin, II, Papadopoulos, KP, Leal, TA, et al. KRYSTAL-1: activity and safety of adagrasib (MRTX849) in patients with colorectal cancer (CRC) and other solid tumors harboring a KRAS G12C mutation. Presented at the 2020 AACR-NCI-EORTC meeting (2020).

  21. Kim D, Xue JY, Lito P. Targeting KRAS(G12C): from inhibitory mechanism to modulation of antitumor effects in patients. Cell. 2020;183:850–9.

    Article  CAS  Google Scholar 

  22. 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. e517

    Article  CAS  Google Scholar 

  23. Patricelli MP, Janes MR, Li LS, Hansen R, Peters U, Kessler LV, et al. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov. 2016;6:316–29.

    Article  CAS  Google Scholar 

  24. Molina-Arcas M, Moore C, Rana S, van Maldegem F, Mugarza E, Romero-Clavijo P, et al. Development of combination therapies to maximize the impact of KRAS-G12C inhibitors in lung cancer. Sci Transl Med. 2019;11:eaaw7999.

    Article  CAS  Google Scholar 

  25. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–5.

    Article  CAS  Google Scholar 

  26. Tiwary P, Limongelli V, Salvalaglio M, Parrinello M. Kinetics of protein–ligand unbinding: predicting pathways, rates, and rate-limiting steps. Proc Natl Acad Sci USA. 2015;112:E386–91.

    Article  CAS  Google Scholar 

  27. Copeland RA. The drug–target residence time model: a 10-year retrospective. Nat Rev Drug Discov. 2016;15:87–95.

    Article  CAS  Google Scholar 

  28. Palkowitz MD, Tan B, Hu H, Roth K, Bauer RA. Synthesis of diverse N-acryloyl azetidines and evaluation of their enhanced thiol reactivities. Org Lett. 2017;19:2270–3.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Dr Hirofumi Ishii, Dr Yukinori Shimoshige and Mr Yosuke Yamanaka for support with the biological study. We are grateful to Dr Masahiko Hayakawa and Dr Taku Yoshida for their useful suggestions.

Funding

This study was funded by Astellas Pharma Inc.

Author information

Authors and Affiliations

Authors

Contributions

AN, TN and MS conceived the study and designed experiments. AN, YN, TN, MS, KK, KM, KH and MY developed methodology. AN, YN, TN, KK, KM, KH and MY acquired and analysed the data. All authors edited the manuscript and approved the submission of the final version.

Corresponding author

Correspondence to Ayako Nakayama.

Ethics declarations

Ethics approval and consent to participate

No human participants, human data or human tissue were used as parts of this study. All animal experimental procedures were approved by the Institutional Animal Care and Use Committee of Astellas Pharma Inc., Tsukuba Research Center, which is accredited by AAALAC International.

Consent for publication

Not applicable.

Competing interests

All authors are employees of Astellas Pharma Inc. and its affiliates.

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

Nakayama, A., Nagashima, T., Nishizono, Y. et al. Characterisation of a novel KRAS G12C inhibitor ASP2453 that shows potent anti-tumour activity in KRAS G12C-mutated preclinical models. Br J Cancer 126, 744–753 (2022). https://doi.org/10.1038/s41416-021-01629-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-021-01629-x

Search

Quick links