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.

  • Resource
  • Published:

Defining the landscape of ATP-competitive inhibitor resistance residues in protein kinases

Abstract

Kinases are involved in disease development and modulation of their activity can be therapeutically beneficial. Drug-resistant mutant kinases are valuable tools in drug discovery efforts, but the prediction of mutants across the kinome is challenging. Here, we generate deep mutational scanning data to identify mutant mammalian kinases that drive resistance to clinically relevant inhibitors. We aggregate these data with subsaturation mutagenesis data and use it to develop, test and validate a framework to prospectively identify residues that mediate kinase activity and drug resistance across the kinome. We validate predicted resistance mutations in CDK4, CDK6, ERK2, EGFR and HER2. Capitalizing on a highly predictable residue, we generate resistance mutations in TBK1, CSNK2A1 and BRAF. Unexpectedly, we uncover a potentially generalizable activation site that mediates drug resistance and confirm its impact in BRAF, EGFR, HER2 and MEK1. We anticipate that the identification of these residues will enable the broad interrogation of the kinome and its inhibitors.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Hotspot analysis of saturating and subsaturating mutagenesis screens identifies common sites that mediate ATP-competitive kinase inhibitor resistance.
Fig. 2: Key common drug-resistant positions mapped onto a ribbon structure of CDK6.
Fig. 3: Mutation of common drug-resistant positions in CDK6, CDK4 and ERK2 causes drug resistance.
Fig. 4: Mutation of common drug-resistant sites differentially impacts drug binding.
Fig. 5: Validation of common drug-resistant positions in EGFR, EML4-ALK, HER2 and BRAF that are associated with drug resistance in patient samples.
Fig. 6: Validation of drug-resistance phenotypes predicted for the Pocket Protector residue (position 32) in TBK1, CSNK2A1 and BRAF.
Fig. 7: Mutation of the Keymaster (position-61) residue leads to gain-of-function activity in protein kinases.

Similar content being viewed by others

Data availability

All previously unpublished saturation mutagenesis data generated or analyzed during this study are included in this published article (and its Extended Data information files). All screening data described in this project can be found in the Supplementary Information or have been deposited in the National Center for Biotechnology Information Short Read Archive (BioProject accession number PRJNA559517). Source data for Fig. 3a,c,e, Fig. 4, Fig. 5a–d,g–j, Fig. 6a,b,d–f, Fig. 7b,d–f, Extended Data Fig. 4a, Extended Data Fig. 6d,e,g and Extended Data Fig. 7c,e are available online. Aggregated mutational scanning data, including previously published datasets, are available from the corresponding author on reasonable request.

Code availability

All code used to analyze mutant screening data (ORFCall) can be accessed at https://github.com/tedsharpe/ORFCall.

References

  1. Daub, H., Specht, K. & Ullrich, A. Strategies to overcome resistance to targeted protein kinase inhibitors. Nat. Rev. Drug Discov. 3, 1001–1010 (2004).

    CAS  PubMed  Google Scholar 

  2. Eyers, P. A., Craxton, M., Morrice, N., Cohen, P. & Goedert, M. Conversion of SB 203580-insensitive MAP kinase family members to drug-sensitive forms by a single amino-acid substitution. Chem. Biol. 5, 321–328 (1998).

    CAS  PubMed  Google Scholar 

  3. Fox, T. et al. A single amino acid substitution makes ERK2 susceptible to pyridinyl imidazole inhibitors of p38 MAP kinase. Protein Sci. 7, 2249–2255 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Liu, Y. et al. Structural basis for selective inhibition of Src family kinases by PP1. Chem. Biol. 6, 671–678 (1999).

    CAS  PubMed  Google Scholar 

  5. Rotow, J. & Bivona, T. G. Understanding and targeting resistance mechanisms in NSCLC. Nat. Rev. Cancer 17, 637–658 (2017).

    CAS  PubMed  Google Scholar 

  6. Whittaker, S. et al. Gatekeeper mutations mediate resistance to BRAF-targeted therapies. Sci. Transl. Med. 2, 35ra41 (2010).

    PubMed  Google Scholar 

  7. Wylie, A. A. et al. The allosteric inhibitor ABL001 enables dual targeting of BCR-ABL1. Nature 543, 733–737 (2017).

    CAS  PubMed  Google Scholar 

  8. Azam, M., Seeliger, M. A., Gray, N. S., Kuriyan, J. & Daley, G. Q. Activation of tyrosine kinases by mutation of the gatekeeper threonine. Nat. Struct. Mol. Biol. 15, 1109–1118 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Carlson, S. M. et al. Large-scale discovery of ERK2 substrates identifies ERK-mediated transcriptional regulation by ETV3. Sci. Signal. 4, rs11 (2011).

    PubMed  PubMed Central  Google Scholar 

  10. Shah, K., Liu, Y., Deirmengian, C. & Shokat, K. M. Engineering unnatural nucleotide specificity for Rous sarcoma virus tyrosine kinase to uniquely label its direct substrates. Proc. Natl Acad. Sci. USA 94, 3565–3570 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Blencke, S., Ullrich, A. & Daub, H. Mutation of threonine 766 in the epidermal growth factor receptor reveals a hotspot for resistance formation against selective tyrosine kinase inhibitors. J. Biol. Chem. 278, 15435–15440 (2003).

    CAS  PubMed  Google Scholar 

  12. Blencke, S. et al. Characterization of a conserved structural determinant controlling protein kinase sensitivity to selective inhibitors. Chem. Biol. 11, 691–701 (2004).

    CAS  PubMed  Google Scholar 

  13. Carter, T. A. et al. Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases. Proc. Natl Acad. Sci. USA 102, 11011–11016 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Gorre, M. E. et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 293, 876–880 (2001).

    CAS  PubMed  Google Scholar 

  15. Kobayashi, S. et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 352, 786–792 (2005).

    CAS  PubMed  Google Scholar 

  16. Pao, W. et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2, e73 (2005).

    PubMed  PubMed Central  Google Scholar 

  17. Shah, N. P. et al. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell 2, 117–125 (2002).

    CAS  PubMed  Google Scholar 

  18. Tamborini, E. et al. A new mutation in the KIT ATP pocket causes acquired resistance to imatinib in a gastrointestinal stromal tumor patient. Gastroenterology 127, 294–299 (2004).

    CAS  PubMed  Google Scholar 

  19. Thomas, R. K. et al. Detection of oncogenic mutations in the EGFR gene in lung adenocarcinoma with differential sensitivity to EGFR tyrosine kinase inhibitors. Cold Spring Harb. Symp. Quant. Biol. 70, 73–81 (2005).

    CAS  PubMed  Google Scholar 

  20. Azam, M., Latek, R. R. & Daley, G. Q. Mechanisms of autoinhibition and STI-571/imatinib resistance revealed by mutagenesis of BCR-ABL. Cell 112, 831–843 (2003).

    CAS  PubMed  Google Scholar 

  21. Emery, C. M. et al. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc. Natl Acad. Sci. USA 106, 20411–20416 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Goetz, E. M., Ghandi, M., Treacy, D. J., Wagle, N. & Garraway, L. A. ERK mutations confer resistance to mitogen-activated protein kinase pathway inhibitors. Cancer Res. 74, 7079–7089 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Lee, B. J. & Shah, N. P. Identification and characterization of activating ABL1 1b kinase mutations: impact on sensitivity to ATP-competitive and allosteric ABL1 inhibitors. Leukemia 31, 1096–1107 (2017).

    CAS  PubMed  Google Scholar 

  24. Marit, M. R. et al. Random mutagenesis reveals residues of JAK2 critical in evading inhibition by a tyrosine kinase inhibitor. PLoS One 7, e43437 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Pahuja, K. B. et al. Actionable Activating Oncogenic ERBB2/HER2 Transmembrane and Juxtamembrane Domain Mutations. Cancer Cell 34, 792–806.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Tiedt, R. et al. A drug resistance screen using a selective MET inhibitor reveals a spectrum of mutations that partially overlap with activating mutations found in cancer patients. Cancer Res. 71, 5255–5264 (2011).

    CAS  PubMed  Google Scholar 

  27. Trowe, T. et al. EXEL-7647 inhibits mutant forms of ErbB2 associated with lapatinib resistance and neoplastic transformation. Clin. Cancer Res. 14, 2465–2475 (2008).

    CAS  PubMed  Google Scholar 

  28. Wagenaar, T. R. et al. Resistance to vemurafenib resulting from a novel mutation in the BRAFV600E kinase domain. Pigment Cell Melanoma Res. 27, 124–133 (2014).

    CAS  PubMed  Google Scholar 

  29. Williams, A. B. et al. Mutations of FLT3/ITD confer resistance to multiple tyrosine kinase inhibitors. Leukemia 27, 48–55 (2013).

    CAS  PubMed  Google Scholar 

  30. Zunder, E. R., Knight, Z. A., Houseman, B. T., Apsel, B. & Shokat, K. M. Discovery of drug-resistant and drug-sensitizing mutations in the oncogenic PI3K isoform p110α. Cancer Cell 14, 180–192 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ahler, E. et al. A combined approach reveals a regulatory mechanism coupling src’s kinase activity, localization, and phosphotransferase-independent functions. Mol. Cell 74, 393–408.e20 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Brenan, L. et al. Phenotypic characterization of a comprehensive set of MAPK1/ERK2 missense mutants. Cell Rep. 17, 1171–1183 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Fowler, D. M. & Fields, S. Deep mutational scanning: a new style of protein science. Nat. Methods 11, 801–807 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Giacomelli, A. O. et al. Mutational processes shape the landscape of TP53 mutations in human cancer. Nat. Genet. 50, 1381–1387 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Gray, V. E., Hause, R. J. & Fowler, D. M. Analysis of large-scale mutagenesis data to assess the impact of single amino acid substitutions. Genetics 207, 53–61 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Gray, V. E., Hause, R. J., Luebeck, J., Shendure, J. & Fowler, D. M. Quantitative missense variant effect prediction using large-scale mutagenesis data. Cell Syst. 6, 116–124.e3 (2018).

    CAS  PubMed  Google Scholar 

  37. Kitzman, J. O., Starita, L. M., Lo, R. S., Fields, S. & Shendure, J. Massively parallel single-amino-acid mutagenesis. Nat. Methods 12, 203–206 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Majithia, A. R. et al. Prospective functional classification of all possible missense variants in PPARG. Nat. Genet. 48, 1570–1575 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Matreyek, K. A. et al. Multiplex assessment of protein variant abundance by massively parallel sequencing. Nat. Genet. 50, 874–882 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Mavor, D. et al. Determination of ubiquitin fitness landscapes under different chemical stresses in a classroom setting. eLife 5, e15802 (2016).

    PubMed  PubMed Central  Google Scholar 

  41. Melnikov, A., Rogov, P., Wang, L., Gnirke, A. & Mikkelsen, T. S. Comprehensive mutational scanning of a kinase in vivo reveals substrate-dependent fitness landscapes. Nucleic Acids Res. 42, e112 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Ray, A., Cowan-Jacob, S. W., Manley, P. W., Mestan, J. & Griffin, J. D. Identification of BCR-ABL point mutations conferring resistance to the Abl kinase inhibitor AMN107 (nilotinib) by a random mutagenesis study. Blood 109, 5011–5015 (2007).

    CAS  PubMed  Google Scholar 

  43. Kolde, R., Laur, S., Adler, P. & Vilo, J. Robust rank aggregation for gene list integration and meta-analysis. Bioinformatics 28, 573–580 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Aronov, A. M. et al. Structure-guided design of potent and selective pyrimidylpyrrole inhibitors of extracellular signal-regulated kinase (ERK) using conformational control. J. Med. Chem. 52, 6362–6368 (2009).

    CAS  PubMed  Google Scholar 

  45. Martinez Molina, D. et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science 341, 84–87 (2013).

    PubMed  Google Scholar 

  46. Yun, C. H. et al. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc. Natl Acad. Sci. USA 105, 2070–2075 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Chen, L., Fu, W., Zheng, L., Liu, Z. & Liang, G. Recent progress of small-molecule epidermal growth factor receptor (EGFR) inhibitors against C797S resistance in non-small-cell lung cancer. J. Med. Chem. 61, 4290–4300 (2018).

    CAS  PubMed  Google Scholar 

  48. Costa, D. B. et al. BIM mediates EGFR tyrosine kinase inhibitor-induced apoptosis in lung cancers with oncogenic EGFR mutations. PLoS Med. 4, 1669–1679 (2007).

    CAS  PubMed  Google Scholar 

  49. Costa, D. B., Schumer, S. T., Tenen, D. G. & Kobayashi, S. Differential responses to erlotinib in epidermal growth factor receptor (EGFR)-mutated lung cancers with acquired resistance to gefitinib carrying the L747S or T790M secondary mutations. J. Clin. Oncol. 26, 1182–1184 (2008).

    PubMed  Google Scholar 

  50. Ercan, D. et al. EGFR mutations and resistance to irreversible pyrimidine-based EGFR inhibitors. Clin. Cancer Res. 21, 3913–3923 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Kohsaka, S. et al. A method of high-throughput functional evaluation of EGFR gene variants of unknown significance in cancer. Sci. Transl. Med. 9, eaan6566 (2017).

    PubMed  Google Scholar 

  52. Liu, Y. et al. Acquired EGFR L718V mutation mediates resistance to osimertinib in non-small cell lung cancer but retains sensitivity to afatinib. Lung Cancer 118, 1–5 (2018).

    PubMed  Google Scholar 

  53. Ou, S. I. et al. Emergence of novel and dominant acquired EGFR solvent-front mutations at Gly796 (G796S/R) together with C797S/R and L792F/H mutations in one EGFR (L858R/T790M) NSCLC patient who progressed on osimertinib. Lung Cancer 108, 228–231 (2017).

    PubMed  Google Scholar 

  54. Wang, Y. T., Ning, W. W., Li, J. & Huang, J. A. Exon 19 L747P mutation presented as a primary resistance to EGFR-TKI: a case report. J. Thorac. Dis. 8, E542–E546 (2016).

    PubMed  PubMed Central  Google Scholar 

  55. Yamaguchi, F. et al. Acquired resistance L747S mutation in an epidermal growth factor receptor-tyrosine kinase inhibitor-naive patient: a report of three cases. Oncol. Lett. 7, 357–360 (2014).

    PubMed  Google Scholar 

  56. Goldberg, M. E. et al. Multiple configurations of EGFR exon 20 resistance mutations after first- and third-generation EGFR TKI treatment affect treatment options in NSCLC. PLoS One 13, e0208097 (2018).

    PubMed  PubMed Central  Google Scholar 

  57. Yang, Z. et al. Investigating novel resistance mechanisms to third-generation EGFR tyrosine kinase inhibitor osimertinib in non-small cell lung cancer patients. Clin. Cancer Res. 24, 3097–3107 (2018).

    CAS  PubMed  Google Scholar 

  58. Bitencourt, R., Zalcberg, I. & Louro, I. D. Imatinib resistance: a review of alternative inhibitors in chronic myeloid leukemia. Rev. Bras. Hematol. Hemoter. 33, 470–475 (2011).

    PubMed  PubMed Central  Google Scholar 

  59. Connor, L. M. O., Langabeer, S., McCann, S. R. & Conneally, E. Mutation mediated resistance to the tyrosine kinase inhibitors imatinib, dasatinib & nilotinib in Philadelphia positive leukaemia. Blood 112, 4245–4245 (2008).

    Google Scholar 

  60. Muller, M. C. et al. Dasatinib treatment of chronic-phase chronic myeloid leukemia: analysis of responses according to preexisting BCR-ABL mutations. Blood 114, 4944–4953 (2009).

    PubMed  PubMed Central  Google Scholar 

  61. Yoda, S. et al. Sequential ALK inhibitors can select for lorlatinib-resistant compound ALK mutations in ALK-positive lung cancer. Cancer Discov. 8, 714–729 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhang, S. et al. The potent ALK inhibitor brigatinib (AP26113) overcomes mechanisms of resistance to first- and second-generation ALK inhibitors in preclinical models. Clin. Cancer Res. 22, 5527–5538 (2016).

    CAS  PubMed  Google Scholar 

  63. Boulbes, D. R. et al. HER family kinase domain mutations promote tumor progression and can predict response to treatment in human breast cancer. Mol. Oncol. 9, 586–600 (2015).

    CAS  PubMed  Google Scholar 

  64. Sun, Z. et al. Analysis of different HER-2 mutations in breast cancer progression and drug resistance. J. Cell. Mol. Med. 19, 2691–2701 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Berger, A. H. et al. High-throughput phenotyping of lung cancer somatic mutations. Cancer Cell 30, 214–228 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Ma, L. et al. CRISPR-Cas9-mediated saturated mutagenesis screen predicts clinical drug resistance with improved accuracy. Proc. Natl Acad. Sci. USA 114, 11751–11756 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Jenkins, R. W. et al. Ex vivo profiling of PD-1 blockade using organotypic tumor spheroids. Cancer Discov. 8, 196–215 (2018).

    CAS  PubMed  Google Scholar 

  68. Zhu, Z. et al. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov. 4, 452–465 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Wu, J. Y. et al. Second-line treatments after first-line gefitinib therapy in advanced nonsmall cell lung cancer. Int. J. Cancer 126, 247–255 (2010).

    CAS  PubMed  Google Scholar 

  70. Wu, J. Y. et al. Effectiveness of tyrosine kinase inhibitors on “uncommon” epidermal growth factor receptor mutations of unknown clinical significance in non-small cell lung cancer. Clin Cancer Res 17, 3812–3821 (2011).

    CAS  PubMed  Google Scholar 

  71. Wu, S. G. et al. Frequent epidermal growth factor receptor gene mutations in malignant pleural effusion of lung adenocarcinoma. Eur. Respir. J. 32, 924–930 (2008).

    CAS  PubMed  Google Scholar 

  72. Kancha, R. K. et al. Differential sensitivity of ERBB2 kinase domain mutations towards lapatinib. PLoS One 6, e26760 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Nagano, M. et al. High-throughput functional evaluation of variants of unknown significance in ERBB2. Clin. Cancer Res. 24, 5112–5122 (2018).

    CAS  PubMed  Google Scholar 

  74. Xu, X. et al. HER2 reactivation through acquisition of the HER2 L755S mutation as a mechanism of acquired resistance to HER2-targeted therapy in HER2+ breast cancer. Clin. Cancer Res. 23, 5123–5134 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Foster, S. A. et al. Activation mechanism of oncogenic deletion mutations in BRAF, EGFR, and HER2. Cancer Cell 29, 477–493 (2016).

    CAS  PubMed  Google Scholar 

  76. Gao, Y. et al. Allele-specific mechanisms of activation of MEK1 mutants determine their properties. Cancer Discov. 8, 648–661 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. He, M. et al. EGFR exon 19 insertions: a new family of sensitizing EGFR mutations in lung adenocarcinoma. Clin. Cancer Res. 18, 1790–1797 (2012).

    CAS  PubMed  Google Scholar 

  78. Kumar, R. D. & Bose, R. Analysis of somatic mutations across the kinome reveals loss-of-function mutations in multiple cancer types. Sci. Rep. 7, 6418 (2017).

    PubMed  PubMed Central  Google Scholar 

  79. Schulze-Gahmen, U., De Bondt, H. L. & Kim, S. H. High-resolution crystal structures of human cyclin-dependent kinase 2 with and without ATP: bound waters and natural ligand as guides for inhibitor design. J. Med. Chem. 39, 4540–4546 (1996).

    CAS  PubMed  Google Scholar 

  80. Thevakumaran, N. et al. Crystal structure of a BRAF kinase domain monomer explains basis for allosteric regulation. Nat. Struct. Mol. Biol. 22, 37–43 (2015).

    CAS  PubMed  Google Scholar 

  81. Zhang, X., Gureasko, J., Shen, K., Cole, P. A. & Kuriyan, J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell 125, 1137–1149 (2006).

    CAS  PubMed  Google Scholar 

  82. Wilson, F. H. et al. A functional landscape of resistance to ALK inhibition in lung cancer. Cancer Cell 27, 397–408 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Yamada, T. et al. Paracrine receptor activation by microenvironment triggers bypass survival signals and ALK inhibitor resistance in EML4-ALK lung cancer cells. Clin. Cancer Res. 18, 3592–3602 (2012).

    CAS  PubMed  Google Scholar 

  84. Nayar, U. et al. Acquired HER2 mutations in ER+ metastatic breast cancer confer resistance to estrogen receptor-directed therapies. Nat. Genet. 51, 207–216 (2019).

    CAS  PubMed  Google Scholar 

  85. Bose, R. et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 3, 224–237 (2013).

    CAS  PubMed  Google Scholar 

  86. Leung, G. P. et al. Hyperactivation of MAPK signaling is deleterious to RAS/RAF-mutant melanoma. Mol. Cancer Res. 17, 199–211 (2019).

    CAS  PubMed  Google Scholar 

  87. Shah, N. H. & Kuriyan, J. Understanding molecular mechanisms in cell signaling through natural and artificial sequence variation. Nat. Struct. Mol. Biol. 26, 25–34 (2019).

    CAS  PubMed  Google Scholar 

  88. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    CAS  PubMed  Google Scholar 

  89. Edgar, R. C. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792–1797 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Klipper-Aurbach, Y. et al. Mathematical formulae for the prediction of the residual beta cell function during the first two years of disease in children and adolescents with insulin-dependent diabetes mellitus. Med. Hypotheses 45, 486–490 (1995).

    CAS  PubMed  Google Scholar 

  91. Jafari, R. et al. The cellular thermal shift assay for evaluating drug target interactions in cells. Nat. Protoc. 9, 2100–2122 (2014).

    CAS  PubMed  Google Scholar 

  92. Haq, R. et al. Oncogenic BRAF regulates oxidative metabolism via PGC1α and MITF. Cancer Cell 23, 302–315 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Burgin, C. Painter, B. Tomson and M. Rees for discussions and critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

N.S.P. and C.M.J. designed the study and wrote the paper. D.H., M.D.C., L.B., O.C., S.K., U.N., A.W., S.P., Y.L., J.C., M.S., C.Z., T.K.H., P.R. and P.P. contributed data for this project. X.Y, C.Z. and N.S.P. analyzed the data, T.S.M., D.A.B., R.H., F.P., D.E.R and C.M.J. supervised the work.

Corresponding author

Correspondence to C. M. Johannessen.

Ethics declarations

Competing interests

C.M.J. is a full-time employee of Novartis Institutes of Biomedical Research, Inc. T.M. is a full-time employee of 10x Genomics.

Additional information

Peer review information Anke Sparmann was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Saturation mutagenesis of CDK4 and CDK6.

a Summary schematic of CDK6 and CDK4 saturating mutagenesis screens. b CDK6 and c CDK4 early time point mutant representation illustrated across the length of each protein. Each dot represents one of two replicate measurements (a and b). d and e Early time point reference correlations for CDK6 and CDK4 mutants. Each dot represents one of two replicate measurements. The pearson correlation score is reported on each graph. f Log(2)-fold-changes for each of four CDK6 replicate measurements was individually calculated with respect to the average of two replicate measurements of early time point mutant representation and shown as black dots. Replicated correlations of Log(2)-Fold-Changes Replicate A versus all three other replicates are shown with their corresponding Pearson correlations. g Log(2)-fold-changes for each of four CDK4 replicate measurements was individually calculated with respect to the average of two replicate measurements of early time point mutant representation and shown as black dots. Replicated correlations of Log(2)-Fold-Changes Replicate A versus all three other replicates are shown with their corresponding Pearson correlations.

Extended Data Fig. 2 Analysis of CDK4/6 saturation mutagenesis data.

a and b Scatter plots of the average Log(2)-Fold-Changes (LFCs, y-axis, n=4 replicate measurements) and native amino acid position (x-axis) for the complete mutagenesis data for CDK6 (a) and CDK4 (b) palbociclib resistance screens. Red lines indicate z-scores of 1.95, our significance threshold for all saturation data (see Methods). Normalized data can be found in Supplementary Tables 1-2. c and d Heatmap visualization of the data presented in (a) and (b) for CDK6 (c) and CDK4 (d). Colors correspond to the z-score of the LFCs. Darker blue indicates a z-score of -3 or lower and darker red indicates a z-score of +3 or higher (more red = more palbociclib resistant).

Extended Data Fig. 3 Spatial projection of CDK4/6 saturation mutagenesis phenotypic data onto the 3D structure of CDK6.

All data for the CDK6 palbociclib resistance screen was reduced to a single metric representing the maximum z-score observed at each position (average of n=4 independent replicates for each of n=19 substitutions). For positions that showed maximum z-scores over 1.95, the z-score was mapped onto the crystal structure of CDK6 bound to palbociclib (PDB 2EUF), either as a ribbon (a) or space-filling diagram (b). c View of the palbociclib binding pocket in CDK6 (PDB 2EUF). For a-c, light orange represents a z-score greater than 1.95 and red represents a z-score of 6 or higher.

Extended Data Fig. 4 Validation of CDK4/6 saturation mutagenesis data.

a Common drug resistant positions found in the CDK6 screen for palbociclib resistance are tested here for resistance to two other CDK4/CDK6 inhibitors: abemaciclib and ribociclib. Average cumulative population doublings after 14 days of Meljuso cells exogenously expressing wild-type (WT) or mutant CDK6 in the absence of drug are shown in turquoise. Treatment with 0.9 µM abemaciclib is shown in light green, and 8 µM ribociclib is shown in dark green, (n=2 independent experiments), error bars represent standard deviation. The correlation of resistance phenotypes in the CDK6 (b) or CDK4 (c) primary screens compared to population doublings in validation experiments, to phosphorylated Rb (compared to no drug) validation experiments, as well as the correlation of validation experiments to each other. Pearson correlations for each set are reported on the graph. Each dot represents a single measurement of the representative western blot shown in main figures.

Source data

Extended Data Fig. 5 Assessment of drug binding to CDK6 mutants using CETSA and DSF.

a Coomasie stained acrylamide gel of purified FLAG-CDK6 mutants used for DSF assays. b DSF assay for investigating the interaction of the drug palbociclib with wild-type and mutant CDK6. The normalized first derivative melt profiles of purified wild-type CDK6, F98E, I19W, V45M, and H100F mutants in the absence (gray trace) or presence (blue trace) of the drug palbociclib. The black arrow indicates the melting profile of CDK6 alone in the absence of the drug. The temperature at which the derivative is the most negative (minimum) is used to calculate the melting temperature (Tm, DTm), average of n = 5 replicate measurements. c CETSA assays of kinase dead CDK6 (K43M) in the presence or absence of palbociclib, average of n=2 replicate measurements.

Extended Data Fig. 6 Confirmation of generalizable kinase mutants beyond CDK4/6.

a WT and mutant EML4-ALK drug sensitivity were measured by Western Blotting of downstream phosphorylation of AKT, representative image of n=2 individual experiments. b Levels of WT and mutant HER2 exogenously expressed in T47D cells were measured by western blotting, representative image of n=2 individual experiments. c Activity of BRAF position 61 (L485S) was measured by western blotting for downstream phosphorylation of MEK and ERK, representative image of n=2 individual experiments. d Bar graphs showing the percentage of all kinases harboring each amino acid at the wild-type residue of position 32, 61 117 and 119. All data is from the kinome alignment described in Kumar, R. D. & Bose, R. (2017). e Bar graphs illustrating the average resistance phenotypes (z-score) caused by all possible amino acid substitutions at the Pocket Protector, Keymaster, Gatekeeper and Gatekeeper +2 residues of CDK4 (n=4 replicate measurements), CDK6 (n=4 replicate measurements) and ERK2 (n=6 replicate measurements), based on CDK4/6 primary screening data and data from Brenan et al. (2016), n= f) Activity of CSNK2A1 mutants as assessed by western blot of downstream signaling, including phosphorylated AKT (pAKT1), representative image of n=2 individual experiments. g Proliferation assay assessing the sensitivity of SKMEL5 cells transduced with WT or mutant CSNK2A1 and treated with CX4945, plotted as the mean of 2 replicate measurements. h Activity of CSNK2A1 mutants as assessed by western blot of downstream signaling, including phosphorylated AKT (pAKT1) corresponding to samples in (g), representative image of n=2 individual experiments.

Source data

Extended Data Fig. 7 Phenotypes of mutant CDK4/6 in standard growth conditions.

Scatter plots of the z-scores of the Log(2)-Fold-Changes and native amino acid position for DMSO (vehicle only, n=2 replicate measurements) calculated relative to the early time point reference for CDK6 (a) or CDK4 (b), n=2 replicate measurements. c Proliferation assay assessing the sensitivity of A375 cells transduced with WT or mutant MEK1 treated with the MEK-inhibitor Trametinib (n=4 replicate measurements for each mutant at each concentration). d Activity of WT or mutant MEK1 as assessed via western blot analysis of downstream signaling, representative blot of n=2 individual experiments. e Dabrafenib complementation assays to measure activity of A375 cells transduced with WT or I99M mutant MEK1 (n=4 replicate measurements for each mutant at each concentration). f Illustration of active and auto-inhibited forms of kinases as illustrated by CDK2 structures (PDB: 2JGZ (active) and PDB: 1HCK (auto-inhibited). Activation loops are colored purple while keymaster positions are colored magenta.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

CDK6 saturation mutagenesis data

Supplementary Table 2

CDK6 saturation mutagenesis data

Source data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 3

Unprocessed Western Blots

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 5

Unprocessed Western Blots

Source Data Fig. 6

Statistical Source Data

Source Data Fig. 6

Unprocessed Western Blots

Source Data Fig. 7

Statistical Source Data

Source Data Fig. 7

Unprocessed Western Blots

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 6

Unprocessed Western Blots

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 7

Unprocessed Western Blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Persky, N.S., Hernandez, D., Do Carmo, M. et al. Defining the landscape of ATP-competitive inhibitor resistance residues in protein kinases. Nat Struct Mol Biol 27, 92–104 (2020). https://doi.org/10.1038/s41594-019-0358-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0358-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing