8azr Citations

Pan-KRAS inhibitor disables oncogenic signalling and tumour growth.

Abstract

KRAS is one of the most commonly mutated proteins in cancer, and efforts to directly inhibit its function have been continuing for decades. The most successful of these has been the development of covalent allele-specific inhibitors that trap KRAS G12C in its inactive conformation and suppress tumour growth in patients1-7. Whether inactive-state selective inhibition can be used to therapeutically target non-G12C KRAS mutants remains under investigation. Here we report the discovery and characterization of a non-covalent inhibitor that binds preferentially and with high affinity to the inactive state of KRAS while sparing NRAS and HRAS. Although limited to only a few amino acids, the evolutionary divergence in the GTPase domain of RAS isoforms was sufficient to impart orthosteric and allosteric constraints for KRAS selectivity. The inhibitor blocked nucleotide exchange to prevent the activation of wild-type KRAS and a broad range of KRAS mutants, including G12A/C/D/F/V/S, G13C/D, V14I, L19F, Q22K, D33E, Q61H, K117N and A146V/T. Inhibition of downstream signalling and proliferation was restricted to cancer cells harbouring mutant KRAS, and drug treatment suppressed KRAS mutant tumour growth in mice, without having a detrimental effect on animal weight. Our study suggests that most KRAS oncoproteins cycle between an active state and an inactive state in cancer cells and are dependent on nucleotide exchange for activation. Pan-KRAS inhibitors, such as the one described here, have broad therapeutic implications and merit clinical investigation in patients with KRAS-driven cancers.

Reviews citing this publication (10)

  1. Precision Oncology: 2023 in Review. Murciano-Goroff YR, Suehnholz SP, Drilon A, Chakravarty D. Cancer Discov 13 2525-2531 (2023)
  2. Prognostic and therapeutic impact of the KRAS G12C mutation in colorectal cancer. Qunaj L, May MS, Neugut AI, Herzberg BO. Front Oncol 13 1252516 (2023)
  3. The current landscape of using direct inhibitors to target KRASG12C-mutated NSCLC. Batrash F, Kutmah M, Zhang J. Exp Hematol Oncol 12 93 (2023)
  4. Current Standards, Multidisciplinary Approaches, and Future Directions in the Management of Extrahepatic Cholangiocarcinoma. Wheless M, Agarwal R, Goff L, Lockney N, Padmanabhan C, Heumann T. Curr Treat Options Oncol (2024)
  5. From bench to bedside: current development and emerging trend of KRAS-targeted therapy. Chen Y, Liu QP, Xie H, Ding J. Acta Pharmacol Sin (2023)
  6. Targeting CRAF kinase in anti-cancer therapy: progress and opportunities. Wang P, Laster K, Jia X, Dong Z, Liu K. Mol Cancer 22 208 (2023)
  7. Targeting KRAS in Colorectal Cancer: A Bench to Bedside Review. Bteich F, Mohammadi M, Li T, Bhat MA, Sofianidi A, Wei N, Kuang C. Int J Mol Sci 24 12030 (2023)
  8. Targeting KRAS in Pancreatic Ductal Adenocarcinoma: The Long Road to Cure. de Jesus VHF, Mathias-Machado MC, de Farias JPF, Aruquipa MPS, Jácome AA, Peixoto RD. Cancers (Basel) 15 5015 (2023)
  9. Targeting KRAS-Mutated Gastrointestinal Malignancies with Small-Molecule Inhibitors: A New Generation of Breakthrough Therapies. Caughey BA, Strickler JH. Drugs 84 27-44 (2024)
  10. The role of CRAF in cancer progression: from molecular mechanisms to precision therapies. Riaud M, Maxwell J, Soria-Bretones I, Dankner M, Li M, Rose AAN. Nat Rev Cancer (2024)

Articles citing this publication (10)

  1. Comment A non-covalent inhibitor with pan-KRAS potential. Kingwell K. Nat Rev Drug Discov 22 622 (2023)
  2. A single inhibitor for all KRAS mutations. Corcoran RB. Nat Cancer 4 1060-1062 (2023)
  3. ASD2023: towards the integrating landscapes of allosteric knowledgebase. He J, Liu X, Zhu C, Zha J, Li Q, Zhao M, Wei J, Li M, Wu C, Wang J, Jiao Y, Ning S, Zhou J, Hong Y, Liu Y, He H, Zhang M, Chen F, Li Y, He X, Wu J, Lu S, Song K, Lu X, Zhang J. Nucleic Acids Res 52 D376-D383 (2024)
  4. Decrypting Allostery in Membrane-Bound K-Ras4B Using Complementary In Silico Approaches Based on Unbiased Molecular Dynamics Simulations. Castelli M, Marchetti F, Osuna S, F Oliveira AS, Mulholland AJ, Serapian SA, Colombo G. J Am Chem Soc 146 901-919 (2024)
  5. Discovery of Hit Compounds Targeting the P4 Allosteric Site of K-RAS, Identified through Ensemble-Based Virtual Screening. Gomez-Gutierrez P, Rubio-Martinez J, Perez JJ. J Chem Inf Model 63 6412-6422 (2023)
  6. Facts and Hopes on RAS Inhibitors and Cancer Immunotherapy. Boumelha J, Molina-Arcas M, Downward J. Clin Cancer Res 29 5012-5020 (2023)
  7. Editorial In the literature: September 2023. Lamarca A, Moreno V, Gambardella V, Cervantes A. ESMO Open 8 102032 (2023)
  8. Mutant KRAS-activated circATXN7 fosters tumor immunoescape by sensitizing tumor-specific T cells to activation-induced cell death. Zhou C, Li W, Liang Z, Wu X, Cheng S, Peng J, Zeng K, Li W, Lan P, Yang X, Xiong L, Zeng Z, Zheng X, Huang L, Fan W, Liu Z, Xing Y, Kang L, Liu H. Nat Commun 15 499 (2024)
  9. Precision Oncology Comes of Age: Designing Best-in-Class Small Molecules by Integrating Two Decades of Advances in Chemistry, Target Biology, and Data Science. Stuart DD, Guzman-Perez A, Brooijmans N, Jackson EL, Kryukov GV, Friedman AA, Hoos A. Cancer Discov 13 2131-2149 (2023)
  10. Stromal-derived NRG1 enables oncogenic KRAS bypass in pancreas cancer. Han J, Xu J, Liu Y, Liang S, LaBella KA, Chakravarti D, Spring DJ, Xia Y, DePinho RA. Genes Dev 37 818-828 (2023)