290
Views
2
CrossRef citations to date
0
Altmetric
Research Articles

Inside the cracked kernel: establishing the molecular basis of AMG510 and MRTX849 in destabilising KRASG12C mutant switch I and II in cancer treatment

ORCID Icon, & ORCID Icon
Pages 4890-4902 | Received 30 Nov 2021, Accepted 30 Apr 2022, Published online: 11 May 2022

References

  • Adjei, A. A., Mauer, A., Bruzek, L., Marks, R. S., Hillman, S., Geyer, S., Hanson, L. J., Wright, J. J., Erlichman, C., Kaufmann, S. H., & Vokes, E. E. (2003). Phase II study of the farnesyl transferase inhibitor R115777 in patients with advanced non-small-cell lung cancer. Journal of Clinical Oncology, 21(9), 1760–1766. https://neuro.unboundmedicine.com/medline/citation/12721252/Phase_II_study_of_the_farnesyl_transferase_inhibitor_R115777_in_patients_with_advanced_non_small_cell_lung_cancer_ https://doi.org/10.1200/JCO.2003.09.075
  • Arora, P., Singh, M., Singh, V., Bhatia, S., & Arora, S. (2021). PROTACs in treatment of cancer: A review. Mini Reviews in Medicinal Chemistry, 21(16), 2347–2360. https://doi.org/10.2174/1389557521666210226150740
  • Berendsen, H. J. C., Postma, J. P. M., van Gunsteren, W. F., DiNola, a., & Haak, J. R. (1984). Molecular dynamics with coupling to an external bath. The Journal of Chemical Physics, 81(8), 3684–3690. https://doi.org/10.1063/1.448118
  • Herman J. C, B., Postma, J. P. M., W. F. van, Hermans, J. G., & van Gunsteren, W. F. (1981). Interaction models for water in relation to protein hydration. Intermolecular Forces, 14, 331–338. https://doi.org/10.1111/j.1574-695X.1996.tb00128.x
  • Berman, H. M., Westbrook, J., Feng, Z., Gilliland, G., Bhat, T. N., Weissig, H., Shindyalov, I. N., & Bourne, P. E. (2000). The Protein Data Bank. Nucleic Acids Research, 28(1), 235–242. https://doi.org/10.1093/nar/28.1.235
  • Bery, N., Miller, A., & Rabbitts, T. (2020). A potent KRAS macromolecule degrader specifically targeting tumours with mutant KRAS. Nature Communications, 11(1), 3233. https://doi.org/10.1038/s41467-020-17022-w
  • BIOVIA, D. S. (2017). Discovery Studio. Dassault Systèmes.
  • Bond, M. J., Chu, L., Nalawansha, D. A., Li, K., & Crews, C. M. (2020). Targeted degradation of oncogenic KRASG12C by VHL-recruiting PROTACs. ACS Central Science, 6(8), 1367–1375. https://doi.org/10.1021/acscentsci.0c00411
  • Bos, J. L. (1989). ras oncogenes in human cancer: A review. Cancer Research, 49(17), 4682–4689.
  • Campbell, J. D., Alexandrov, A., Kim, J., Wala, J., Berger, A. H., Pedamallu, C. S., Shukla, S. A., Guo, G., Brooks, A. N., Murray, B. A., Imielinski, M., Hu, X., Ling, S., Akbani, R., Rosenberg, M., Cibulskis, C., Ramachandran, A., Collisson, E. A., Kwiatkowski, D. J., … Meyerson, M., Cancer Genome Atlas Research Network. (2016). Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nature Genetics, 48(6), 607–616. https://doi.org/10.1038/ng.3564
  • Canon, J., Rex, K., Saiki, A. Y., Mohr, C., Cooke, K., Bagal, D., Gaida, K., Holt, T., Knutson, C. G., Koppada, N., Lanman, B. A., Werner, J., Rapaport, A. S., San Miguel, T., Ortiz, R., Osgood, T., Sun, J.-R., Zhu, X., McCarter, J. D., … Lipford, J. R. (2019). The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature, 575(7781), 217–223. https://doi.org/10.1038/s41586-019-1694-1
  • Case, D. A., Babin, V., Berryman, J., Betz, R. M., Cai, Q., Cerutti, D. S., Cheatham, T. E., Darden, T. A., Duke, R. E., Roe, D. R., LeGrand, S., Swails, J., Gotz, A. W., Smith, J., Ceerutti, D., Brozell, S. R., Luchko, T., Cruzeiro, V. W. D., Ghoreishi, D., … Gohlke, H. (2018). Amber 18. San Francisco: University of California.
  • Chamberlain, P. P., & Cathers, B. E. (2019). Cereblon modulators: Low molecular weight inducers of protein degradation. Drug Discovery Today. Technologies, 31, 29–34. https://doi.org/10.1016/j.ddtec.2019.02.004
  • Chen, H., Smaill, J. B., Liu, T., Ding, K., & Lu, X. (2020). Small-molecule inhibitors directly targeting KRAS as anticancer therapeutics. Journal of Medicinal Chemistry, 63(23), 14404–14424. https://doi.org/10.1021/acs.jmedchem.0c01312
  • Cisowski, J., Sayin, V. I., Liu, M., Karlsson, C., & Bergo, M. O. (2016). Oncogene-induced senescence underlies the mutual exclusive nature of oncogenic KRAS and BRAF. Oncogene, 35(10), 1328–1333. https://doi.org/10.1038/onc.2015.186
  • Cisowski, J., & Bergo, M. O. (2017). What makes oncogenes mutually exclusive? Small GTPases, 8(3), 187–192. https://doi.org/10.1080/21541248.2016.1212689
  • Cox, A. D., Fesik, S. W., Kimmelman, A. C., Luo, J., & Der, C. J. (2014). Drugging the undruggable RAS: Mission possible? Nature Reviews. Drug Discovery, 13(11), 828–851. https://doi.org/10.1038/nrd4389
  • Daviter, T., Johnson, C. M., & Mclaughlin, S. H, Eds. (2021). Protein-ligand interactions. Springer sciences. https://doi.org/10.1007/978-1-0716-1197-5
  • Fell, J. B., Fischer, J. P., Baer, B. R., Blake, J. F., Bouhana, K., Briere, D. M., Brown, K. D., Burgess, L. E., Burns, A. C., Burkard, M. R., Chiang, H., Chicarelli, M. J., Cook, A. W., Gaudino, J. J., Hallin, J., Hanson, L., Hartley, D. P., Hicken, E. J., Hingorani, G. P., … Marx, M. A. (2020). Identification of the clinical development candidate MRTX849, a covalent KRASG12C inhibitor for the treatment of cancer. Journal of Medicinal Chemistry, 63 (13), 6679–6693. https://doi.org/10.1021/acs.jmedchem.9b02052
  • Fell, J. B., Fischer, J. P., Baer, B. R., Ballard, J., Blake, J. F., Bouhana, K., Brandhuber, B. J., Briere, D. M., Burgess, L. E., Burkard, M. R., Chiang, H., Chicarelli, M. J., Davidson, K., Gaudino, J. J., Hallin, J., Hanson, L., Hee, K., Hicken, E. J., Hinklin, R. J., … Christensen, J. G. (2018). Discovery of tetrahydropyridopyrimidines as irreversible covalent inhibitors of KRAS-G12C with in vivo activity. ACS Medicinal Chemistry Letters, 9(12), 1230–1234. https://doi.org/10.1021/acsmedchemlett.8b00382
  • Genheden, S., & Ryde, U. (2015). The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities. Expert Opinion on Drug Discovery, 10(5), 449–461. https://doi.org/10.1517/17460441.2015.1032936
  • Gonnet, P. (2007). P-SHAKE: A quadratically convergent SHAKE in O (n2). Journal of Computational Physics, 220(2), 740–750. https://doi.org/10.1016/j.jcp.2006.05.032
  • Herdeis, L., Gerlach, D., McConnell, D. B., & Kessler, D. (2021). Stopping the beating heart of cancer: KRAS reviewed. Current Opinion in Structural Biology, 71, 136–147. https://doi.org/10.1016/j.sbi.2021.06.013
  • Heymach, J. V., Johnson, D. H., Khuri, F. R., Safran, H., Schlabach, L. L., Yunus, F., DeVore, R. F., De Porre, P. M., Richards, H. M., Jia, X., Zhang, S., & Johnson, B. E. (2004). Phase II study of the farnesyl transferase inhibitor R115777 in patients with sensitive relapse small-cell lung cancer. Annals of Oncology, 15(8), 1187–1193. https://doi.org/10.1093/annonc/mdh315
  • Hobbs, G. A., Der, C. J., & Rossman, K. L. (2016). RAS isoforms and mutations in cancer at a glance. Journal of Cell Science, 129(7), 1287–1292. https://doi.org/10.1242/jcs.182873
  • Hospital, A., Goñi, J. R., Orozco, M., & Gelpí, J. L. (2015). Molecular dynamics simulations: Advances and applications. Advances and Applications in Bioinformatics and Chemistry, 37. https://doi.org/10.2147/AABC.S70333
  • Humphrey, W., Dalke, A., & Schulten, K. (1996). VMD: Visual molecular dynamics. Journal of Molecular Graphics, 14(1), 33–28. https://doi.org/10.1016/0263-7855(96)00018-5
  • Janes, M. R., Zhang, J., Li, L.-S., Hansen, R., Peters, U., Guo, X., Chen, Y., Babbar, A., Firdaus, S. J., Darjania, L., Feng, J., Chen, J. H., Li, S., Li, S., Long, Y. O., Thach, C., Liu, Y., Zarieh, A., Ely, T., … Liu, Y. (2018). Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell, 172(3), 578–589.e17. https://doi.org/10.1016/j.cell.2018.01.006
  • Kettle, J. G., Bagal, S. K., Bickerton, S., Bodnarchuk, M. S., Breed, J., Carbajo, R. J., Cassar, D. J., Chakraborty, A., Cosulich, S., Cumming, I., Davies, M., Eatherton, A., Evans, L., Feron, L., Fillery, S., Gleave, E. S., Goldberg, F. W., Harlfinger, S., Hanson, L., … Zhao, X. (2020). Structure-based design and pharmacokinetic optimization of covalent allosteric inhibitors of the mutant GTPase KRASG12C). Journal of Medicinal Chemistry, 63(9), 4468–4483. https://doi.org/10.1021/acs.jmedchem.9b01720
  • Khan, S., Bjij, I., Betz, R. M., & Soliman, M. E. S. (2018). Reversible versus irreversible inhibition modes of ERK2: A comparative analysis for ERK2 protein kinase in cancer therapy. Future Medicinal Chemistry, 10(9), 1003–1015. https://doi.org/10.4155/fmc-2017-0275
  • Koča, J., Svobodová, R., Lukáš Pravda, V., Berka, K., Geidl, S., Sehnal, D., & Otyepka, M. (2016). Springer briefs in biochemistry and molecular biology. Structural bioinformatics tools for drug design extraction of biologically relevant information from structural databases. http://www.springer.com/series/10196
  • Krönke, J., Udeshi, N. D., Narla, A., Grauman, P., Hurst, S. N., McConkey, M., Svinkina, T., Heckl, D., Comer, E., Li, X., Ciarlo, C., Hartman, E., Munshi, N., Schenone, M., Schreiber, S. L., Carr, S. A., & Ebert, B. L. (2014). Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science (New York, NY), 343(6168), 301–305. https://doi.org/10.1126/science.1244851
  • Langer, T., & Hoffmann, R. D. (2006). Pharmacophore modelling: applications in drug discovery. Expert Opinion on Drug Discovery, 1(3), 261–267. https://doi.org/10.1517/17460441.1.3.261
  • Lanman, B. A., Allen, J. R., Allen, J. G., Amegadzie, A. K., Ashton, K. S., Booker, S. K., Chen, J. J., Chen, N., Frohn, M. J., Goodman, G., Kopecky, D. J., Liu, L., Lopez, P., Low, J. D., Ma, V., Minatti, A. E., Nguyen, T. T., Nishimura, N., Pickrell, A. J., … Cee, V. J. (2020). Discovery of a covalent inhibitor of KRASG12C (AMG 510) for the treatment of solid tumors. Journal of Medicinal Chemistry, 63(1), 52–65. https://doi.org/10.1021/acs.jmedchem.9b01180
  • Lito, P., Solomon, M., Li, L.-S., Hansen, R., & Rosen, N. (2016). Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science (New York, N.Y.), 351(6273), 604–608. https://doi.org/10.1126/science.aad6204
  • Madhavi Sastry, G., Adzhigirey, M., Day, T., Annabhimoju, R., & Sherman, W. (2013). Protein and ligand preparation: Parameters, protocols, and influence on virtual screening enrichments. Journal of Computer-Aided Molecular Design, 27(3), 221–234.
  • Maier, J. A., Martinez, C., Kasavajhala, K., Wickstrom, L., Hauser, K. E., & Simmerling, C. (2015). ff14SB: Improving the accuracy of protein side chain and backbone parameters from ff99SB. Journal of Chemical Theory and Computation, 11(8), 3696–3713. https://doi.org/10.1021/acs.jctc.5b00255
  • Maiorov, V. N., & Crippen, G. M. (1994). Significance of root-mean-square deviation in comparing three-dimensional structures of globular proteins. Journal of Molecular Biology, 235(2), 625–634. https://doi.org/10.1006/jmbi.1994.1017
  • Matyskiela, M. E., Couto, S., Zheng, X., Lu, G., Hui, J., Stamp, K., Drew, C., Ren, Y., Wang, M., Carpenter, A., Lee, C.-W., Clayton, T., Fang, W., Lu, C.-C., Riley, M., Abdubek, P., Blease, K., Hartke, J., Kumar, G., … Chamberlain, P. P. (2018). SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nature Chemical Biology, 14(10), 981–987. https://doi.org/10.1038/s41589-018-0129-x
  • McDonald, E. R., de Weck, A., Schlabach, M. R., Billy, E., Mavrakis, K. J., Hoffman, G. R., Belur, D., Castelletti, D., Frias, E., Gampa, K., Golji, J., Kao, I., Li, L., Megel, P., Perkins, T. A., Ramadan, N., Ruddy, D. A., Silver, S. J., Sovath, S., … Sellers, W. R. (2017). Project DRIVE: A compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening. Cell, 170(3), 577–592.e10. https://doi.org/10.1016/j.cell.2017.07.005
  • McGee, J. H., Shim, S. Y., Lee, S.-J., Swanson, P. K., Jiang, S. Y., Durney, M. A., & Verdine, G. L. (2018). Exceptionally high-affinity Ras binders that remodel its effector domain. The Journal of Biological Chemistry, 293(9), 3265–3280. https://doi.org/10.1074/jbc.M117.816348
  • Miller, B. R., Mcgee, T. D., Swails, J. M., Homeyer, N., Gohlke, H., & Roitberg, A. E. (2012). MMPBSA. py: An efficient program for end-state free energy calculations. https://doi.org/10.1021/ct300418h
  • Mullard, A. (2019). Cracking KRAS. Nature Reviews. Drug Discovery, 18(12), 887–891. https://doi.org/10.1038/d41573-019-00195-5
  • Nnadi, C. I., Jenkins, M. L., Gentile, D. R., Bateman, L. A., Zaidman, D., Balius, T. E., Nomura, D. K., Burke, J. E., Shokat, K. M., & London, N. (2018). Novel K-Ras G12C switch-II covalent binders destabilize ras and accelerate nucleotide exchange. Journal of Chemical Information and Modeling, 58(2), 464–471. https://doi.org/10.1021/acs.jcim.7b00399
  • Ostrem, J. M. L., & Shokat, K. M. (2016). Direct small-molecule inhibitors of KRAS: From structural insights to mechanism-based design. Nature Reviews. Drug Discovery, 15(11), 771–785. https://doi.org/10.1038/nrd.2016.139
  • Ostrem, J. M., Peters, U., Sos, M. L., Wells, J. A., & Shokat, K. M. (2013). K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature, 503(7477), 548–551. https://doi.org/10.1038/nature12796
  • Pantsar, T. (2020). The current understanding of KRAS protein structure and dynamics. Computational and Structural Biotechnology Journal, 18, 189–198. https://doi.org/10.1016/j.csbj.2019.12.004
  • Patricelli, M. P., Janes, M. R., Li, L.-S., Hansen, R., Peters, U., Kessler, L. V., Chen, Y., Kucharski, J. M., Feng, J., Ely, T., Chen, J. H., Firdaus, S. J., Babbar, A., Ren, P., & Liu, Y. (2016). Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discovery, 6(3), 316–329. https://doi.org/10.1158/2159-8290.CD-15-1105
  • Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S., Greenblatt, D. M., Meng, E. C., & Ferrin, T. E. (2004). UCSF Chimera-A visualization system for exploratory research and analysis. Journal of Computational Chemistry, 25(13), 1605–1612. https://doi.org/10.1002/jcc.20084
  • Phase 3 study of MRTX849 with cetuximab vs. chemotherapy in patients with advanced colorectal cancer with KRAS G12C mutation (KRYSTAL-10) | smart patients. (2021.). https://www.smartpatients.com/trials/NCT04793958
  • Raha, K., & Merz, K. M. (2005). Chapter 9 calculating binding free energy in protein-ligand interaction. Annual Reports in Computational Chemistry, 1(C), 113–130. https://doi.org/10.1016/S1574-1400(05)01009-1
  • Rastelli, G., Del Rio, A., Degliesposti, G., & Sgobba, M. (2010). Fast and accurate predictions of binding free energies using MM-PBSA and MM-GBSA. Journal of Computational Chemistry, 31(4), 797–810. https://doi.org/10.1002/jcc.21372
  • Rehman, I., Farooq, M., & Botelho, S. (2020). Biochemistry, secondary protein structure. StatPearls. https://www.ncbi.nlm.nih.gov/books/NBK470235/
  • Roe, D. R., & Cheatham, T. E. (2013). PTRAJ and CPPTRAJ: Software for processing and analysis of molecular dynamics trajectory data. Journal of Chemical Theory and Computation, 9(7), 3084–3095. https://doi.org/10.1021/ct400341p
  • Seifert, E. (2014). OriginPro 9.1: Scientific data analysis and graphing software-software review. Journal of Chemical Information and Modeling, 54(5), 1552–1552. https://doi.org/10.1021/ci500161d
  • Simanshu, D. K., Nissley, D. V., & McCormick, F. (2017). RAS proteins and their regulators in human disease. Cell, 170(1), 17–33. https://doi.org/10.1016/j.cell.2017.06.009
  • Lu, J., Harrison, R. A., Li, L., Zeng, M., Gondi, S., Scott, D., Gray, N. S., Engen, J. R., & Westover, K. D. (2017). KRAS G12C drug development: Discrimination between switch II pocket configurations using hydrogen/deuterium-exchange mass spectrometry. Structure (London, England : 1993), 25(9), 1442–1448.e3. https://doi.org/10.1016/j.str.2017.07.003
  • States, U., Partnership, D., & Biopharma, A. (2018). Direct targeting of Kras mutant cancers with a Kras G12C mutation-selective inhibitor the Kras revival (pp. 1–9).
  • Study to Compare AMG 510 ‘Proposed INN Sotorasib’ with docetaxel in non small cell lung cancer (NSCLC) (CodeBreak 200). - full text view – ClinicalTrials. (2021). https://www.clinicaltrials.gov/ct2/show/NCT04303780
  • Tong, L., de Vos, A. M., Milburn, M. V., & Kim, S.-H. (1991). Crystal structures at 2.2 Å resolution of the catalytic domains of normal ras protein and an oncogenic mutant complexed with GDP. Journal of Molecular Biology, 217(3), 503–516. https://doi.org/10.1016/0022-2836(91)90753-S
  • Tsherniak, A., Vazquez, F., Montgomery, P. G., Weir, B. A., Kryukov, G., Cowley, G. S., Gill, S., Harrington, W. F., Pantel, S., Krill-Burger, J. M., Meyers, R. M., Ali, L., Goodale, A., Lee, Y., Jiang, G., Hsiao, J., Gerath, W. F. J., Howell, S., Merkel, E., … Hahn, W. C. (2017). Defining a cancer dependency map. Cell, 170(3), 564–576.e16. https://doi.org/10.1016/j.cell.2017.06.010
  • Wang, X., Allen, S., Blake, J. F., Bowcut, V., Briere, D. M., Calinisan, A., … Marx, M. A. (2021). Identification of MRTX1133, a noncovalent, potent, and selective KRAS. Journal of Medicinal Chemistry, 65(4):3123–3133. https://doi.org/10.1021/acs.jmedchem.1c01688
  • Waters, A. M., & Der, C. J. (2018). KRAS: The critical driver and therapeutic target for pancreatic cancer. Cold Spring Harbor Perspectives in Medicine, 8(9), a031435–18. https://doi.org/10.1101/cshperspect.a031435
  • Wu, Y., Tong, J., Ding, K., Zhou, Q., & Zhao, S. (2019). Protein allostery in drug discovery. Advances in Experimental Medicine and Biology, 1163, 25–45. https://doi.org/10.1007/978-981-13-8719-7_10
  • Xie, M., Xu, X., & Fan, Y. (2021). KRAS-mutant non-small cell lung cancer: an emerging promisingly treatable subgroup. Frontiers in Oncology, 11, 672612. https://doi.org/10.3389/fonc.2021.672612
  • Zehir, A., Benayed, R., Shah, R. H., Syed, A., Middha, S., Kim, H. R., Srinivasan, P., Gao, J., Chakravarty, D., Devlin, S. M., Hellmann, M. D., Barron, D. A., Schram, A. M., Hameed, M., Dogan, S., Ross, D. S., Hechtman, J. F., DeLair, D. F., Yao, J., … Berger, M. F. (2017). Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nature Medicine, 23(6), 703–713. https://doi.org/10.1038/nm.4333
  • Zeng, M., Xiong, Y., Safaee, N., Nowak, R. P., Donovan, K. A., Yuan, C. J., Nabet, B., Gero, T. W., Feru, F., Li, L., Gondi, S., Ombelets, L. J., Quan, C., Jänne, P. A., Kostic, M., Scott, D. A., Westover, K. D., Fischer, E. S., & Gray, N. S. (2020). Exploring targeted degradation strategy for oncogenic KRASG12C). Cell Chemical Biology, 27(1), 19–31.e6. https://doi.org/10.1016/j.chembiol.2019.12.006
  • Zhang, Z., & Shokat, K. M. (2019). Bifunctional small-molecule ligands of K-Ras induce its association with immunophilin proteins. Angewandte Chemie (International ed. in English), 58(45), 16314–16319. https://doi.org/10.1002/anie.201910124

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.