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.

  • Review Article
  • Published:

Trends in kinase drug discovery: targets, indications and inhibitor design

An Author Correction to this article was published on 02 September 2021

This article has been updated

Abstract

The FDA approval of imatinib in 2001 was a breakthrough in molecularly targeted cancer therapy and heralded the emergence of kinase inhibitors as a key drug class in the oncology area and beyond. Twenty years on, this article analyses the landscape of approved and investigational therapies that target kinases and trends within it, including the most popular targets of kinase inhibitors and their expanding range of indications. There are currently 71 small-molecule kinase inhibitors (SMKIs) approved by the FDA and an additional 16 SMKIs approved by other regulatory agencies. Although oncology is still the predominant area for their application, there have been important approvals for indications such as rheumatoid arthritis, and one-third of the SMKIs in clinical development address disorders beyond oncology. Information on clinical trials of SMKIs reveals that approximately 110 novel kinases are currently being explored as targets, which together with the approximately 45 targets of approved kinase inhibitors represent only about 30% of the human kinome, indicating that there are still substantial unexplored opportunities for this drug class. We also discuss trends in kinase inhibitor design, including the development of allosteric and covalent inhibitors, bifunctional inhibitors and chemical degraders.

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: Timeline of approved kinase inhibitors.
Fig. 2: FDA-approved kinase inhibitors mapped onto the human kinome.
Fig. 3: Structural features of the kinase catalytic domain and inhibitor binding modes.
Fig. 4: Chemical scaffolds used in approved kinase inhibitors.
Fig. 5: Schematic representation of kinase inhibitor binding modes.
Fig. 6: Exploring the kinome.
Fig. 7: Disease indications of kinase-targeting agents in clinical trials.

Similar content being viewed by others

Data availability

Data are provided in the Supplementary information. Supplementary Table 1 provides information on protein kinase inhibitors approved by selected regulatory agencies (FDA, EMA, NMPA, PMDA and MFDS) and their primary therapeutic targets. Supplementary Table 2 provides information on agents in clinical trials that target protein kinase signalling pathways. Supplementary Table 3 provides success rates of FDA-approved small-molecule kinase inhibitors in clinical trials. Supplementary Table 4 provides information on kinase targets for which modulators in clinical trials are presumed discontinued. Supplementary Table 5 provides information on chemical probes and historical compounds for kinases. Supplementary Fig. 1 provides information on active and not active small-molecule kinase inhibitors targeting established and novel kinases.

Change history

References

  1. Deribe, Y. L., Pawson, T. & Dikic, I. Post-translational modifications in signal integration. Nat. Struct. Mol. Biol. 17, 666–672 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Ardito, F., Giuliani, M., Perrone, D., Troiano, G. & Lo Muzio, L. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (Review). Int. J. Mol. Med. 40, 271–280 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kanev, G. K. et al. The landscape of atypical and eukaryotic protein kinases. Trends Pharmacol. Sci. 40, 818–832 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Manning, G., Whyte, D. B., Martinez, R., Hunter, T. & Sudarsanam, S. The protein kinase complement of the human genome. Science 298, 1912–1934 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Parsons, S. J. & Parsons, J. T. Src family kinases, key regulators of signal transduction. Oncogene 23, 7906–7909 (2004).

    Article  CAS  PubMed  Google Scholar 

  6. Tamaoki, T. et al. Staurosporine, a potent inhibitor of phospholipid/Ca++ dependent protein kinase. Biochem. Biophys. Res. Commun. 135, 397–402 (1986).

    Article  CAS  PubMed  Google Scholar 

  7. Sun, L. et al. Synthesis and biological evaluations of 3-substituted indolin-2-ones: a novel class of tyrosine kinase inhibitors that exhibit selectivity toward particular receptor tyrosine kinases. J. Med. Chem. 41, 2588–2603 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. Barker, A. J. et al. Studies leading to the identification of ZD1839 (IRESSA): an orally active, selective epidermal growth factor receptor tyrosine kinase inhibitor targeted to the treatment of cancer. Bioorg. Med. Chem. Lett. 11, 1911–1914 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Zimmermann, J. et al. Phenylamino-pyrimidine (PAP) derivatives: a new class of potent and selective inhibitors of protein kinase C (PKC). Arch. Pharm. Weinh. 329, 371–376 (1996).

    Article  CAS  Google Scholar 

  10. Heitman, J., Movva, N. R. & Hall, M. N. Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science 253, 905–909 (1991).

    Article  CAS  PubMed  Google Scholar 

  11. Kunz, J. et al. Target of rapamycin in yeast, TOR2, is an essential phosphatidylinositol kinase homolog required for G1 progression. Cell 73, 585–596 (1993).

    Article  CAS  PubMed  Google Scholar 

  12. Brown, E. J. et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 369, 756–758 (1994).

    Article  CAS  PubMed  Google Scholar 

  13. Sabatini, D. M., Erdjument-Bromage, H., Lui, M., Tempst, P. & Snyder, S. H. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 78, 35–43 (1994).

    Article  CAS  PubMed  Google Scholar 

  14. Pfizer. Rapamune (sirolimus) oral solution. Prescribing Information. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021083s067,021110s085lbl.pdf (FDA, 2020).

  15. Jürg Zimmermann. Interview with Jürg Zimmermann, global head of oncology & exploratory chemistry at Novartis. Future Med. Chem. 1, 1395–1398 (2009).

    Article  PubMed  Google Scholar 

  16. Fabbro, D. in Inhibitors of Protein Kinases and Protein Phosphates (eds Pinna, L. A. & Cohen, P. T. W.) 361–389 (Springer, 2005).

  17. Zhang, Q., Zheng, P. & Zhu, W. Research progress of small molecule VEGFR/c-met inhibitors as anticancer agents (2016-present). Molecules 25, 2666 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  18. Huang, L., Jiang, S. & Shi, Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020). J. Hematol. Oncol. J. Hematol. Oncol. 13, 143 (2020).

    Article  PubMed  CAS  Google Scholar 

  19. Wu, P., Nielsen, T. E. & Clausen, M. H. FDA-approved small-molecule kinase inhibitors. Trends Pharmacol. Sci. 36, 422–439 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Wu, P., Nielsen, T. E. & Clausen, M. H. Small-molecule kinase inhibitors: an analysis of FDA-approved drugs. Drug Discov. Today 21, 5–10 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Szilveszter, K. P., Németh, T. & Mócsai, A. Tyrosine kinases in autoimmune and inflammatory skin diseases. Front. Immunol. 10, 1862 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Xie, Z., Yang, X., Duan, Y., Han, J. & Liao, C. Small-molecule kinase inhibitors for the treatment of nononcologic diseases. J. Med. Chem. 64, 1283–1345 (2021).

    Article  CAS  PubMed  Google Scholar 

  23. Yang, J. et al. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol. Cancer 18, 26 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Roskoski, R. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol. Res. 103, 26–48 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Roskoski, R. Properties of FDA-approved small molecule protein kinase inhibitors: a 2021 update. Pharmacol. Res. 165, 105463 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Fabbro, D., Cowan-Jacob, S. W. & Moebitz, H. Ten things you should know about protein kinases: IUPHAR Review 14. Br. J. Pharmacol. 172, 2675–2700 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Fedorov, O., Müller, S. & Knapp, S. The (un)targeted cancer kinome. Nat. Chem. Biol. 6, 166–169 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Roskoski, R. A historical overview of protein kinases and their targeted small molecule inhibitors. Pharmacol. Res. 100, 1–23 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Stransky, N., Cerami, E., Schalm, S., Kim, J. L. & Lengauer, C. The landscape of kinase fusions in cancer. Nat. Commun. 5, 4846 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Lahiry, P., Torkamani, A., Schork, N. J. & Hegele, R. A. Kinase mutations in human disease: interpreting genotype–phenotype relationships. Nat. Rev. Genet. 11, 60–74 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Fleuren, E. D., Zhang, L., Wu, J. & Daly, R. J. The kinome ‘at large’ in cancer. Nat. Rev. Cancer 16, 83–98 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Essegian, D., Khurana, R., Stathias, V. & Schürer, S. C. The clinical kinase index: a method to prioritize understudied kinases as drug targets for the treatment of cancer. Cell Rep. Med. 1, 100128 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tuna, M., Amos, C. I. & Mills, G. B. Molecular mechanisms and pathobiology of oncogenic fusion transcripts in epithelial tumors. Oncotarget 10, 2095–2111 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Ben-Neriah, Y., Daley, G. Q., Mes-Masson, A. M., Witte, O. N. & Baltimore, D. The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene. Science 233, 212–214 (1986).

    Article  CAS  PubMed  Google Scholar 

  35. Jabbour, E. & Kantarjian, H. Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am. J. Hematol. 95, 691–709 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Liu, J. et al. Recent advances in Bcr-Abl tyrosine kinase inhibitors for overriding T315I mutation. Chem. Biol. Drug Des. 97, 649–664 (2020).

    Article  PubMed  CAS  Google Scholar 

  37. Chen, K.-K., Du, T.-F., Xiong, P.-S., Fan, G.-H. & Yang, W. Discontinuation of tyrosine kinase inhibitors in chronic myeloid leukemia with losing major molecular response as a definition for molecular relapse: a systematic review and meta-analysis. Front. Oncol. 9, 372 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Cerveira, N. et al. Discontinuation of tyrosine kinase inhibitors in CML patients in real-world clinical practice at a single institution. BMC Cancer 18, 1245 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. American Cancer Society. Lung cancer statistics. How common is lung cancer. Cancer.org https://www.cancer.org/cancer/lung-cancer/about/key-statistics.html (2021).

  40. de Groot, P. M., Wu, C. C., Carter, B. W. & Munden, R. F. The epidemiology of lung cancer. Transl Lung Cancer Res. 7, 220–233 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Neel, D. S. & Bivona, T. G. Resistance is futile: overcoming resistance to targeted therapies in lung adenocarcinoma. NPJ Precis. Oncol. 1, 1–6 (2017).

    Google Scholar 

  42. Vu, P. & Patel, S. P. Non-small cell lung cancer targetable mutations: present and future. Precis. Cancer Med. https://doi.org/10.21037/pcm.2019.11.03 (2020).

    Article  Google Scholar 

  43. Guo, Y. et al. Recent progress in rare oncogenic drivers and targeted therapy for non-small cell lung cancer. OncoTargets Ther. 12, 10343–10360 (2019).

    Article  CAS  Google Scholar 

  44. Brabender, J. et al. Epidermal growth factor receptor and HER2-neu mRNA expression in non-small cell lung cancer is correlated with survival. Clin. Cancer Res. 7, 1850–1855 (2001).

    CAS  PubMed  Google Scholar 

  45. Ricordel, C., Friboulet, L., Facchinetti, F. & Soria, J.-C. Molecular mechanisms of acquired resistance to third-generation EGFR-TKIs in EGFR T790M-mutant lung cancer. Ann. Oncol. 29, i28–i37 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Gazdar, A. Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 28, S24–S31 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lynch, T. J. et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non–small-cell lung cancer to gefitinib. N. Engl. J. Med. 350, 2129–2139 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Kuan, F.-C. et al. Overall survival benefits of first-line EGFR tyrosine kinase inhibitors in EGFR-mutated non-small-cell lung cancers: a systematic review and meta-analysis. Br. J. Cancer 113, 1519–1528 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Vyse, S. & Huang, P. H. Targeting EGFR exon 20 insertion mutations in non-small cell lung cancer. Signal. Transduct. Target. Ther. 4, 1–10 (2019).

    Google Scholar 

  50. Leonetti, A. et al. Resistance mechanisms to osimertinib in EGFR -mutated non-small cell lung cancer. Br. J. Cancer 121, 725–737 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Kazandjian, D. et al. FDA approval summary: crizotinib for the treatment of metastatic non-small cell lung cancer with anaplastic lymphoma kinase rearrangements. Oncologist 19, e5–e11 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Bergethon, K. et al. ROS1 rearrangements define a unique molecular class of lung cancers. J. Clin. Oncol. 30, 863–870 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gainor, J. F. et al. Patterns of metastatic spread and mechanisms of resistance to crizotinib in ROS1-positive non–small-cell lung cancer. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00063 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Della Corte, C. M. et al. Role and targeting of anaplastic lymphoma kinase in cancer. Mol. Cancer 17, 30 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. FDA. FDA approves selpercatinib for lung and thyroid cancers with RET gene mutations or fusions. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-selpercatinib-lung-and-thyroid-cancers-ret-gene-mutations-or-fusions (2020).

  56. FDA. FDA approves pralsetinib for lung cancer with RET gene fusions. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pralsetinib-lung-cancer-ret-gene-fusions (2020).

  57. FDA. FDA grants accelerated approval to capmatinib for metastatic non-small cell lung cancer. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-capmatinib-metastatic-non-small-cell-lung-cancer (2020).

  58. FDA. FDA grants accelerated approval to tepotinib for metastatic non-small cell lung cancer. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-tepotinib-metastatic-non-small-cell-lung-cancer (2021).

  59. Buti, S., Leonetti, A., Dallatomasina, A. & Bersanelli, M. Everolimus in the management of metastatic renal cell carcinoma: an evidence-based review of its place in therapy. Core Evid. 11, 23–36 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Abhinand, C. S., Raju, R., Soumya, S. J., Arya, P. S. & Sudhakaran, P. R. VEGF-A/VEGFR2 signaling network in endothelial cells relevant to angiogenesis. J. Cell Commun. Signal. 10, 347–354 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Dagher, R. et al. Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clin. Cancer Res. 8, 3034–3038 (2002).

    CAS  PubMed  Google Scholar 

  62. Mei, L. et al. Gastrointestinal stromal tumors: the GIST of precision medicine. Trends Cancer 4, 74–91 (2018).

    Article  CAS  PubMed  Google Scholar 

  63. Abou-Alfa, G. K. et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 21, 671–684 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. FDA. FDA approves first targeted treatment patients cholangiocarcinoma cancer bile ducts. FDA.gov https://www.fda.gov/news-events/press-announcements/fda-approves-first-targeted-treatment-patients-cholangiocarcinoma-cancer-bile-ducts (2020).

  65. Xuhong, J.-C., Qi, X.-W., Zhang, Y. & Jiang, J. Mechanism, safety and efficacy of three tyrosine kinase inhibitors lapatinib, neratinib and pyrotinib in HER2-positive breast cancer. Am. J. Cancer Res. 9, 2103–2119 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Sánchez-Martínez, C., Lallena, M. J., Sanfeliciano, S. G. & de Dios, A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015–2019). Bioorg. Med. Chem. Lett. 29, 126637 (2019).

    Article  PubMed  CAS  Google Scholar 

  67. FDA. FDA approves drug to reduce bone marrow suppression caused by chemotherapy. FDA.gov https://www.fda.gov/news-events/press-announcements/fda-approves-drug-reduce-bone-marrow-suppression-caused-chemotherapy (2021).

  68. Domingues, B., Lopes, J. M., Soares, P. & Pópulo, H. Melanoma treatment in review. ImmunoTargets Ther. 7, 35–49 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. FDA. FDA approves dabrafenib plus trametinib for adjuvant treatment of melanoma with BRAF V600E or V600K mutations. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-dabrafenib-plus-trametinib-adjuvant-treatment-melanoma-braf-v600e-or-v600k-mutations (2018).

  70. Yu, C. et al. Combination of immunotherapy with targeted therapy: theory and practice in metastatic melanoma. Front. Immunol. 10, 990 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ugurel, S. et al. Survival of patients with advanced metastatic melanoma: the impact of novel therapies–update 2017. Eur. J. Cancer 83, 247–257 (2017).

    Article  PubMed  Google Scholar 

  72. Yang, Q., Modi, P., Newcomb, T., Queva, C. & Gandhi, V. Idelalisib: first-in-class PI3K delta inhibitor for the treatment of chronic lymphocytic leukemia, small lymphocytic leukemia, and follicular lymphoma. Clin. Cancer Res. 21, 1537–1542 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Gilead Sciences. ZYDELIG (idelalisib) tablets, for oral use. Prescribing Information. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/205858s014lbl.pdf (FDA, 2020).

  74. FDA. FDA grants accelerated approval to copanlisib for relapsed follicular lymphoma. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-copanlisib-relapsed-follicular-lymphoma (2017).

  75. FDA. duvelisib (COPIKTRA, Verastem, Inc.) for adult patients with relapsed or refractory chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL). FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/duvelisib-copiktra-verastem-inc-adult-patients-relapsed-or-refractory-chronic-lymphocytic-leukemia (2018).

  76. FDA. FDA approves alpelisib for metastatic breast cancer. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-alpelisib-metastatic-breast-cancer (2019).

  77. Burger, J. A. Bruton tyrosine kinase inhibitors: present and future. Cancer J. Sudbury Mass. 25, 386–393 (2019).

    Article  CAS  Google Scholar 

  78. Looney, A.-M., Nawaz, K. & Webster, R. M. Tumour-agnostic therapies. Nat. Rev. Drug Discov. 19, 383–384 (2020).

    Article  CAS  PubMed  Google Scholar 

  79. Martin-Zanca, D., Hughes, S. H. & Barbacid, M. A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature 319, 743–748 (1986).

    Article  CAS  PubMed  Google Scholar 

  80. FDA. FDA approves larotrectinib for solid tumors with NTRK gene fusions. FDA.gov https://www.fda.gov/drugs/fda-approves-larotrectinib-solid-tumors-ntrk-gene-fusions (2019).

  81. FDA. FDA approves entrectinib for NTRK solid tumors and ROS-1 NSCLC. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-entrectinib-ntrk-solid-tumors-and-ros-1-nsclc (2019).

  82. Banerjee, S., Biehl, A., Gadina, M., Hasni, S. & Schwartz, D. M. JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs 77, 521–546 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zarrin, A. A., Bao, K., Lupardus, P. & Vucic, D. Kinase inhibition in autoimmunity and inflammation. Nat. Rev. Drug Discov. 20, 39–63 (2021).

    Article  CAS  PubMed  Google Scholar 

  84. Vainchenker, W. & Constantinescu, S. N. A unique activating mutation in JAK2 (V617F) is at the origin of polycythemia vera and allows a new classification of myeloproliferative diseases. Hematol. Am. Soc. Hematol. Educ. Program https://doi.org/10.1182/asheducation-2005.1.195 (2005).

    Article  Google Scholar 

  85. Takada, Y. & Aggarwal, B. B. TNF activates syk protein tyrosine kinase leading to TNF-induced MAPK activation, NF-κB activation, and apoptosis. J. Immunol. 173, 1066–1077 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Crowley, M. T. et al. A critical role for syk in signal transduction and phagocytosis mediated by Fcγ receptors on macrophages. J. Exp. Med. 186, 1027–1039 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Mócsai, A., Ruland, J. & Tybulewicz, V. L. J. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat. Rev. Immunol. 10, 387–402 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Mullard, A. FDA approves first-in-class SYK inhibitor. Nat. Rev. Drug. Discov. 17, 385–385 (2018).

    PubMed  Google Scholar 

  89. FDA. FDA approves everolimus for tuberous sclerosis complex-associated partial-onset seizures. FDA.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-everolimus-tuberous-sclerosis-complex-associated-partial-onset-seizures (2018).

  90. McCormack, P. L. Nintedanib: first global approval. Drugs 75, 129–139 (2015).

    Article  CAS  PubMed  Google Scholar 

  91. Kasamon, Y. L. et al. FDA approval summary: midostaurin for the treatment of advanced systemic mastocytosis. Oncologist 23, 1511–1519 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hoy, S. M. Netarsudil ophthalmic solution 0.02%: first global approval. Drugs 78, 389–396 (2018).

    Article  CAS  PubMed  Google Scholar 

  93. Smolinski, M. P. et al. Discovery of novel dual mechanism of action src signaling and tubulin polymerization inhibitors (KX2-391 and KX2-361). J. Med. Chem. 61, 4704–4719 (2018).

    Article  CAS  PubMed  Google Scholar 

  94. Lombardo, L. J. et al. Discovery of N-(2-chloro-6-methyl- phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4- ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J. Med. Chem. 47, 6658–6661 (2004).

    Article  CAS  PubMed  Google Scholar 

  95. Motzer, R. J., Hoosen, S., Bello, C. L. & Christensen, J. G. Sunitinib malate for the treatment of solid tumours: a review of current clinical data. Expert. Opin. Investig. Drugs 15, 553–561 (2006).

    Article  CAS  PubMed  Google Scholar 

  96. Faivre, S., Demetri, G., Sargent, W. & Raymond, E. Molecular basis for sunitinib efficacy and future clinical development. Nat. Rev. Drug Discov. 6, 734–745 (2007).

    Article  CAS  PubMed  Google Scholar 

  97. Cheng, H. & Force, T. Why do kinase inhibitors cause cardiotoxicity and what can be done about it? Prog. Cardiovasc. Dis. 53, 114–120 (2010).

    Article  CAS  PubMed  Google Scholar 

  98. Odogwu, L. et al. FDA approval summary: dabrafenib and trametinib for the treatment of metastatic non-small cell lung cancers harboring BRAF V600E mutations. Oncologist 23, 740–745 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Yu, S. et al. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Exp. Hematol. Oncol. 6, 31 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Knighton, D. R. et al. Structure of a peptide inhibitor bound to the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase. Science 253, 414–420 (1991).

    Article  CAS  PubMed  Google Scholar 

  101. Knighton, D. R. et al. Crystal structure of the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase. Science 253, 407–414 (1991).

    Article  CAS  PubMed  Google Scholar 

  102. Schindler, T. et al. Structural mechanism for STI-571 inhibition of abelson tyrosine kinase. Science 289, 1938–1942 (2000).

    Article  CAS  PubMed  Google Scholar 

  103. Nolen, B., Taylor, S. & Ghosh, G. Regulation of protein kinases; controlling activity through activation segment conformation. Mol. Cell 15, 661–675 (2004).

    Article  CAS  PubMed  Google Scholar 

  104. Kornev, A. P., Haste, N. M., Taylor, S. S. & Eyck, L. F. T. Surface comparison of active and inactive protein kinases identifies a conserved activation mechanism. Proc. Natl Acad. Sci. Usa. 103, 17783–17788 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Kornev, A. P., Taylor, S. S. & Ten Eyck, L. F. A helix scaffold for the assembly of active protein kinases. Proc. Natl Acad. Sci. USA 105, 14377–14382 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Meharena, H. S. et al. Deciphering the structural basis of eukaryotic protein kinase regulation. PLoS Biol. 11, e1001680 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  109. 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).

    Article  CAS  PubMed  Google Scholar 

  110. Rudolf, A. F., Skovgaard, T., Knapp, S., Jensen, L. J. & Berthelsen, J. A comparison of protein kinases inhibitor screening methods using both enzymatic activity and binding affinity determination. PLOS ONE 9, e98800 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Fedorov, O., Niesen, F. H. & Knapp, S. in Kinase Inhibitors: Methods and Protocols (ed. Kuster, B.) 109–118 (Humana Press, 2012).

  112. Karaman, M. W. et al. A quantitative analysis of kinase inhibitor selectivity. Nat. Biotechnol. 26, 127–132 (2008).

    Article  CAS  PubMed  Google Scholar 

  113. Bantscheff, M. et al. Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat. Biotechnol. 25, 1035–1044 (2007).

    Article  CAS  PubMed  Google Scholar 

  114. Drewes, G. & Knapp, S. Chemoproteomics and chemical probes for target discovery. Trends Biotechnol. 36, 1275–1286 (2018).

    Article  CAS  PubMed  Google Scholar 

  115. Robers, M. B. et al. Quantifying target occupancy of small molecules within living cells. Annu. Rev. Biochem. 89, 557–581 (2020).

    Article  CAS  PubMed  Google Scholar 

  116. Vasta, J. D. et al. Quantitative, wide-spectrum kinase profiling in live cells for assessing the effect of cellular ATP on target engagement. Cell Chem. Biol. 25, 206–214.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Georgi, V. et al. Binding kinetics survey of the drugged kinome. J. Am. Chem. Soc. 140, 15774–15782 (2018).

    Article  CAS  PubMed  Google Scholar 

  118. Berger, B.-T. et al. Structure-kinetic relationship reveals the mechanism of selectivity of FAK inhibitors over PYK2. Cell Chem. Biol. https://doi.org/10.1016/j.chembiol.2021.01.003 (2021).

    Article  PubMed  Google Scholar 

  119. Kanev, G. K., de Graaf, C., Westerman, B. A., de Esch, I. J. P. & Kooistra, A. J. KLIFS: an overhaul after the first 5 years of supporting kinase research. Nucleic Acids Res. 49, D562–D569 (2021).

    Article  CAS  PubMed  Google Scholar 

  120. Müller, S., Chaikuad, A., Gray, N. S. & Knapp, S. The ins and outs of selective kinase inhibitor development. Nat. Chem. Biol. 11, 818–821 (2015).

    Article  PubMed  CAS  Google Scholar 

  121. Guimarães, C. R. W. et al. Understanding the impact of the P-loop conformation on kinase selectivity. J. Chem. Inf. Model. 51, 1199–1204 (2011).

    Article  PubMed  CAS  Google Scholar 

  122. Zuccotto, F., Ardini, E., Casale, E. & Angiolini, M. Through the ‘gatekeeper door’: exploiting the active kinase conformation. J. Med. Chem. 53, 2681–2694 (2010).

    Article  CAS  PubMed  Google Scholar 

  123. Waizenegger, I. C. et al. A novel RAF kinase inhibitor with DFG-Out-binding mode: high efficacy in BRAF-mutant tumor xenograft models in the absence of normal tissue hyperproliferation. Mol. Cancer Ther. 15, 354–365 (2016).

    Article  CAS  PubMed  Google Scholar 

  124. Wood, E. R. et al. A unique structure for epidermal growth factor receptor bound to GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Res. 64, 6652–6659 (2004).

    Article  CAS  PubMed  Google Scholar 

  125. Axten, J. M. et al. Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). J. Med. Chem. 55, 7193–7207 (2012).

    Article  CAS  PubMed  Google Scholar 

  126. Wang, H., Blais, J., Ron, D. & Cardozo, T. Structural determinants of PERK inhibitor potency and selectivity. Chem. Biol. Drug Des. 76, 480–495 (2010).

    Article  CAS  PubMed  Google Scholar 

  127. Chaikuad, A. et al. A unique inhibitor binding site in ERK1/2 is associated with slow binding kinetics. Nat. Chem. Biol. 10, 853–860 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Buchanan, S. G. et al. SGX523 is an exquisitely selective, ATP-competitive inhibitor of the MET receptor tyrosine kinase with antitumor activity in vivo. Mol. Cancer Ther. 8, 3181–3190 (2009).

    Article  CAS  PubMed  Google Scholar 

  129. Kooistra, A. J. et al. KLIFS: a structural kinase-ligand interaction database. Nucleic Acids Res. 44, D365–D371 (2016).

    Article  CAS  PubMed  Google Scholar 

  130. Kohlmann, A., Zhu, X. & Dalgarno, D. Application of MM-GB/SA and watermap to src kinase inhibitor potency prediction. ACS Med. Chem. Lett. 3, 94–99 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Cyphers, S., Ruff, E. F., Behr, J. M., Chodera, J. D. & Levinson, N. M. A water-mediated allosteric network governs activation of Aurora kinase A. Nat. Chem. Biol. 13, 402–408 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Tomita, N. et al. Structure-based discovery of cellular-active allosteric inhibitors of FAK. Bioorg. Med. Chem. Lett. 23, 1779–1785 (2013).

    Article  CAS  PubMed  Google Scholar 

  133. Bagal, S. K. et al. Discovery of allosteric, potent, subtype selective, and peripherally restricted TrkA kinase inhibitors. J. Med. Chem. 62, 247–265 (2019).

    Article  CAS  PubMed  Google Scholar 

  134. Karpov, A. S. et al. Optimization of a dibenzodiazepine hit to a potent and selective allosteric PAK1 Inhibitor. ACS Med. Chem. Lett. 6, 776–781 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Heinrich, T., Grädler, U., Böttcher, H., Blaukat, A. & Shutes, A. Allosteric IGF-1R inhibitors. ACS Med. Chem. Lett. 1, 199–203 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Zhang, J. et al. Targeting Bcr–Abl by combining allosteric with ATP-binding-site inhibitors. Nature 463, 501–506 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Nagar, B. et al. Structural basis for the autoinhibition of c-Abl tyrosine kinase. Cell 112, 859–871 (2003).

    Article  CAS  PubMed  Google Scholar 

  138. Hughes, T. P. et al. Asciminib in chronic myeloid leukemia after ABL kinase inhibitor failure. N. Engl. J. Med. 381, 2315–2326 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Huang, B. X. et al. Identification of 4-phenylquinolin-2(1H)-one as a specific allosteric inhibitor of Akt. Sci. Rep. 7, 11673 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Hirai, H. et al. MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol. Cancer Ther. 9, 1956–1967 (2010).

    Article  CAS  PubMed  Google Scholar 

  141. Rettenmaier, T. J. et al. A small-molecule mimic of a peptide docking motif inhibits the protein kinase PDK1. Proc. Natl Acad. Sci. USA 111, 18590–18595 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Jia, Y. et al. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature 534, 129–132 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Dungo, R. T. & Keating, G. M. Afatinib: first global approval. Drugs 73, 1503–1515 (2013).

    Article  CAS  PubMed  Google Scholar 

  144. Serafimova, I. M. et al. Reversible targeting of noncatalytic cysteines with chemically tuned electrophiles. Nat. Chem. Biol. 8, 471–476 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Bradshaw, J. M. et al. Prolonged and tunable residence time using reversible covalent kinase inhibitors. Nat. Chem. Biol. 11, 525–531 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Forster, M. et al. Selective JAK3 inhibitors with a covalent reversible binding mode targeting a new induced fit binding pocket. Cell Chem. Biol. 23, 1335–1340 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Chaikuad, A., Koch, P., Laufer, S. A. & Knapp, S. The cysteinome of protein kinases as a target in drug development. Angew. Chem. Int. Ed. Engl. 57, 4372–4385 (2018).

    Article  CAS  PubMed  Google Scholar 

  148. Rao, S. et al. Leveraging compound promiscuity to identify targetable cysteines within the kinome. Cell Chem. Biol. 26, 818–829 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Johnson, T. W. et al. Discovery of (10R)-7-Amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a macrocyclic inhibitor of anaplastic lymphoma kinase (ALK) and c-ros oncogene 1 (ROS1) with preclinical brain exposure and broad-spectrum potency against ALK-resistant mutations. J. Med. Chem. 57, 4720–4744 (2014).

    Article  CAS  PubMed  Google Scholar 

  150. Bauer, T. M. et al. Brain penetration of lorlatinib: cumulative incidences of CNS and Non-CNS progression with lorlatinib in patients with previously treated ALK-positive non-small-cell lung cancer. Target. Oncol. 15, 55–65 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  151. Zou, H. Y. et al. PF-06463922 is a potent and selective next-generation ROS1/ALK inhibitor capable of blocking crizotinib-resistant ROS1 mutations. Proc. Natl Acad. Sci. USA 112, 3493–3498 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Engelhardt, H. et al. Start selective and rigidify: the discovery path toward a next generation of EGFR tyrosine kinase inhibitors. J. Med. Chem. 62, 10272–10293 (2019).

    Article  CAS  PubMed  Google Scholar 

  153. Gower, C. M., Chang, M. E. K. & Maly, D. J. Bivalent inhibitors of protein kinases. Crit. Rev. Biochem. Mol. Biol. 49, 102–115 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Cox, K. J., Shomin, C. D. & Ghosh, I. Tinkering outside the kinase ATP box: allosteric (type IV) and bivalent (type V) inhibitors of protein kinases. Future Med. Chem. 3, 29–43 (2011).

    Article  CAS  PubMed  Google Scholar 

  155. Johnson, T. K. & Soellner, M. B. Bivalent inhibitors of c-Src tyrosine kinase that bind a regulatory domain. Bioconjug. Chem. 27, 1745–1749 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Ekambaram, R. et al. Selective bisubstrate inhibitors with sub-nanomolar affinity for protein kinase Pim-1. ChemMedChem 8, 909–913 (2013).

    Article  CAS  PubMed  Google Scholar 

  157. Burslem, G. M. & Crews, C. M. Proteolysis-targeting chimeras as therapeutics and tools for biological discovery. Cell 181, 102–114 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Lai, A. C. & Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discov. 16, 101–114 (2017).

    Article  CAS  PubMed  Google Scholar 

  159. Donovan, K. A. et al. Mapping the degradable kinome provides a resource for expedited degrader development. Cell 183, 1714–1731 (2020).

    Article  CAS  PubMed  Google Scholar 

  160. Posternak, G. et al. Functional characterization of a PROTAC directed against BRAF mutant V600E. Nat. Chem. Biol. 16, 1170–1178 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Zhang, X. et al. Design and synthesis of selective degraders of EGFRL858R/T790M mutant. Eur. J. Med. Chem. 192, 112199 (2020).

    Article  CAS  PubMed  Google Scholar 

  162. Shah, R. R. et al. Hi-JAK-ing the ubiquitin system: The design and physicochemical optimisation of JAK PROTACs. Bioorg. Med. Chem. 28, 115326 (2020).

    Article  CAS  PubMed  Google Scholar 

  163. Tinworth, C. P. et al. PROTAC-mediated degradation of Bruton’s tyrosine kinase is inhibited by covalent binding. ACS Chem. Biol. 14, 342–347 (2019).

    Article  CAS  PubMed  Google Scholar 

  164. Zhang, C. et al. Proteolysis targeting chimeras (PROTACs) of anaplastic lymphoma kinase (ALK). Eur. J. Med. Chem. 151, 304–314 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. You, I. et al. Discovery of an AKT degrader with prolonged inhibition of downstream signaling. Cell Chem. Biol. 27, 66–73 (2020).

    Article  CAS  PubMed  Google Scholar 

  166. Brand, M. et al. Homolog-selective degradation as a strategy to probe the function of CDK6 in AML. Cell Chem. Biol. 26, 300–306.e9 (2019).

    Article  CAS  PubMed  Google Scholar 

  167. Wei, M. et al. First orally bioavailable prodrug of proteolysis targeting chimera (PROTAC) degrades cyclin-dependent kinases 2/4/6 in vivo. Eur. J. Med. Chem. 209, 112903 (2021).

    Article  CAS  PubMed  Google Scholar 

  168. Robb, C. M. et al. Chemically induced degradation of CDK9 by a proteolysis targeting chimera (PROTAC). Chem. Commun. Camb. Engl. 53, 7577–7580 (2017).

    Article  CAS  Google Scholar 

  169. Crew, A. P. et al. Identification and characterization of von Hippel-Lindau-recruiting proteolysis targeting chimeras (PROTACs) of TANK-binding kinase 1. J. Med. Chem. 61, 583–598 (2018).

    Article  CAS  PubMed  Google Scholar 

  170. Adhikari, B. et al. PROTAC-mediated degradation reveals a non-catalytic function of AURORA-A kinase. Nat. Chem. Biol. 16, 1179–1188 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Xiang, W. & Wang, S. Selectively targeting tropomyosin receptor kinase a (TRKA) via PROTACs. J. Med. Chem. 63, 14560–14561 (2020).

    Article  CAS  PubMed  Google Scholar 

  172. Gao, H. et al. Design, synthesis, and evaluation of highly potent FAK-targeting PROTACs. ACS Med. Chem. Lett. 11, 1855–1862 (2020).

    Article  CAS  PubMed  Google Scholar 

  173. Mullard, A. First targeted protein degrader hits the clinic. Nat. Rev. Drug Discov. 18, 237–239 (2019).

    Google Scholar 

  174. Chen, J., Zheng, X. F., Brown, E. J. & Schreiber, S. L. Identification of an 11-kDa FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue. Proc. Natl Acad. Sci. USA 92, 4947–4951 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Ito, T. et al. Identification of a primary target of thalidomide teratogenicity. Science 327, 1345–1350 (2010).

    Article  CAS  PubMed  Google Scholar 

  176. Matyskiela, M. E. et al. SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nat. Chem. Biol. 14, 981–987 (2018).

    Article  CAS  PubMed  Google Scholar 

  177. Krönke, J. et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305 (2014).

    Article  PubMed  CAS  Google Scholar 

  178. Petzold, G., Fischer, E. S. & Thomä, N. H. Structural basis of lenalidomide-induced CK1α degradation by the CRL4(CRBN) ubiquitin ligase. Nature 532, 127–130 (2016).

    Article  CAS  PubMed  Google Scholar 

  179. Dubost, C. Primary hyperparathyroidism: the surgical problems. A study of 1,300 operated patients. Horm. Res. 32, 101–103 (1989).

    Article  CAS  PubMed  Google Scholar 

  180. Słabicki, M. et al. The CDK inhibitor CR8 acts as a molecular glue degrader that depletes cyclin K. Nature 585, 293–297 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Rask-Andersen, M., Zhang, J., Fabbro, D. & Schiöth, H. B. Advances in kinase targeting: current clinical use and clinical trials. Trends Pharmacol. Sci. 35, 604–620 (2014).

    Article  CAS  PubMed  Google Scholar 

  182. Hauser, A. S., Attwood, M. M., Rask-Andersen, M., Schiöth, H. B. & Gloriam, D. E. Trends in GPCR drug discovery: new agents, targets and indications. Nat. Rev. Drug Discov. 16, 829–842 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Hay, M., Thomas, D. W., Craighead, J. L., Economides, C. & Rosenthal, J. Clinical development success rates for investigational drugs. Nat. Biotechnol. 32, 40–51 (2014).

    Article  CAS  PubMed  Google Scholar 

  184. DiMasi, J. A. & Grabowski, H. G. Economics of new oncology drug development. J. Clin. Oncol. 8, 209–216 (2007).

    Article  Google Scholar 

  185. DiMasi, J. A., Feldman, L., Seckler, A. & Wilson, A. Trends in risks associated with new drug development: success rates for investigational drugs. Clin. Pharmacol. Ther. 87, 272–277 (2010).

    Article  CAS  PubMed  Google Scholar 

  186. FDA. Drug development process: clinical research. FDA.org https://www.fda.gov/patients/learn-about-drug-and-device-approvals/drug-development-process (2019).

  187. Walker, I. & Newell, H. Do molecularly targeted agents in oncology have reduced attrition rates? Nat. Rev. Drug Discov. 8, 15–16 (2009).

    Article  CAS  PubMed  Google Scholar 

  188. Morgan, P. et al. Can the flow of medicines be improved? Fundamental pharmacokinetic and pharmacological principles toward improving phase II survival. Drug Discov. Today 17, 419–424 (2012).

    Article  CAS  PubMed  Google Scholar 

  189. Morgan, P. et al. Impact of a five-dimensional framework on R&D productivity at AstraZeneca. Nat. Rev. Drug Discov. 17, 167–181 (2018).

    Article  CAS  PubMed  Google Scholar 

  190. Venot, Q. et al. Targeted therapy in patients with PIK3CA-related overgrowth syndrome. Nature 558, 540–546 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Hillmann, P. & Fabbro, D. PI3K/mTOR pathway inhibition: opportunities in oncology and rare genetic diseases. Int. J. Mol. Sci. 20, 5792 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  192. Rao, V. K. et al. Effective ‘activated PI3Kδ syndrome’-targeted therapy with the PI3Kδ inhibitor leniolisib. Blood 130, 2307–2316 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Sandborn, W. J. et al. Development of gut-selective pan-Janus kinase inhibitor TD-1473 for ulcerative colitis: a translational medicine programme. J. Crohns Colitis 14, 1202–1213 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  194. Montalban, X. et al. Placebo-controlled trial of an oral BTK inhibitor in multiple sclerosis. N. Engl. J. Med. 380, 2406–2417 (2019).

    Article  CAS  PubMed  Google Scholar 

  195. Haselmayer, P. et al. Efficacy and pharmacodynamic modeling of the BTK inhibitor evobrutinib in autoimmune disease models. J. Immunol. 202, 2888–2906 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Carmena, M., Earnshaw, W. C. & Glover, D. M. The dawn of aurora kinase research: from fly genetics to the clinic. Front. Cell Dev. Biol. 3, 73 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  197. Bavetsias, V. & Linardopoulos, S. Aurora kinase inhibitors: current status and outlook. Front. Oncol. 5, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  198. Asghar, U., Witkiewicz, A. K., Turner, N. C. & Knudsen, E. S. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat. Rev. Drug Discov. 14, 130–146 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Li, L., Guan, Y., Chen, X., Yang, J. & Cheng, Y. DNA repair pathways in cancer therapy and resistance. Front. Pharmacol. 11, 629266 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Rundle, S., Bradbury, A., Drew, Y. & Curtin, N. J. Targeting the ATR-CHK1 axis in cancer therapy. Cancers Basel 9, 41 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  201. Dinarello, C. A. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood 117, 3720–3732 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Degterev, A., Ofengeim, D. & Yuan, J. Targeting RIPK1 for the treatment of human diseases. Proc. Natl Acad. Sci. USA 116, 9714–9722 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Harris, P. A. et al. Discovery of a first-in-class receptor interacting protein 1 (RIP1) kinase specific clinical candidate (GSK2982772) for the treatment of inflammatory diseases. J. Med. Chem. 60, 1247–1261 (2017).

    Article  CAS  PubMed  Google Scholar 

  204. Gantke, T., Sriskantharajah, S. & Ley, S. C. Regulation and function of TPL-2, an IκB kinase-regulated MAP kinase kinase kinase. Cell Res. 21, 131–145 (2011).

    Article  CAS  PubMed  Google Scholar 

  205. Senger, K. et al. The kinase TPL2 activates ERK and p38 signaling to promote neutrophilic inflammation. Sci. Signal. 10, eaah4273 (2017).

    Article  PubMed  CAS  Google Scholar 

  206. Sriskantharajah, S. et al. Regulation of experimental autoimmune encephalomyelitis by TPL-2 kinase. J. Immunol. 192, 3518–3529 (2014).

    Article  CAS  PubMed  Google Scholar 

  207. Liu, C. et al. Discovery of BMS-986202: a clinical Tyk2 inhibitor that binds to Tyk2 JH2. J. Med. Chem. 64, 677–694 (2021).

    Article  CAS  PubMed  Google Scholar 

  208. Papp, K. et al. Phase 2 trial of selective tyrosine kinase 2 inhibition in psoriasis. N. Engl. J. Med. 379, 1313–1321 (2018).

    Article  CAS  PubMed  Google Scholar 

  209. Deshmukh, V. et al. A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee. Osteoarthr. Cartil. 26, 18–27 (2018).

    Article  CAS  Google Scholar 

  210. Deshmukh, V. et al. Modulation of the Wnt pathway through inhibition of CLK2 and DYRK1A by lorecivivint as a novel, potentially disease-modifying approach for knee osteoarthritis treatment. Osteoarthr. Cartil. 27, 1347–1360 (2019).

    Article  CAS  Google Scholar 

  211. Yazici, Y. et al. Lorecivivint, a novel intraarticular CDC-like kinase 2 and dual-specificity tyrosine phosphorylation-regulated kinase 1A inhibitor and Wnt pathway modulator for the treatment of knee osteoarthritis: a phase II randomized trial. Arthritis Rheumatol. 72, 1694–1706 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Lee, J. C. et al. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature 372, 739–746 (1994).

    Article  CAS  PubMed  Google Scholar 

  213. Scior, T., Domeyer, D. M., Cuanalo-Contreras, K. & Laufer, S. A. Pharmacophore design of p38α MAP kinase inhibitors with either 2,4,5-trisubstituted or 1,2,4,5-tetrasubstituted imidazole scaffold. Curr. Med. Chem. 18, 1526–1539 (2011).

    Article  CAS  PubMed  Google Scholar 

  214. Dambach, D. M. Potential adverse effects associated with inhibition of p38α/β MAP kinases. Curr. Top. Med. Chem. 5, 929–939 (2005).

    Article  CAS  PubMed  Google Scholar 

  215. Regan, J. et al. The kinetics of binding to p38MAP kinase by analogues of BIRB 796. Bioorg. Med. Chem. Lett. 13, 3101–3104 (2003).

    Article  CAS  PubMed  Google Scholar 

  216. Hammaker, D. & Firestein, G. S. “Go upstream, young man”: lessons learned from the p38 saga. Ann. Rheum. Dis. 69, i77–i82 (2010).

    Article  CAS  PubMed  Google Scholar 

  217. Haller, V., Nahidino, P., Forster, M. & Laufer, S. A. An updated patent review of p38 MAP kinase inhibitors (2014-2019). Expert Opin. Ther. Pat. 30, 453–466 (2020).

    Article  CAS  PubMed  Google Scholar 

  218. Asih, P. R. et al. Functions of p38 MAP kinases in the central nervous system. Front. Mol. Neurosci. 13, 570586 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Duffy, J. P. et al. The discovery of VX-745: a novel and selective p38α kinase inhibitor. ACS Med. Chem. Lett. 2, 758–763 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Heneka, M. T. et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 14, 388–405 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Nygaard, H. B. Targeting fyn kinase in Alzheimer’s disease. Biol. Psychiatry 83, 369–376 (2018).

    Article  CAS  PubMed  Google Scholar 

  222. Ron, D. & Berger, A. Targeting the intracellular signaling ‘STOP’ and ‘GO’ pathways for the treatment of alcohol use disorders. Psychopharmacol. Berl. 235, 1727–1743 (2018).

    Article  CAS  Google Scholar 

  223. West, A. B. et al. Parkinson’s disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc. Natl Acad. Sci. USA 102, 16842–16847 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Daher, J. P. L. et al. Leucine-rich repeat kinase 2 (LRRK2) pharmacological inhibition abates α-synuclein gene-induced neurodegeneration. J. Biol. Chem. 290, 19433–19444 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Kostich, W. et al. Inhibition of AAK1 kinase as a novel therapeutic approach to treat neuropathic pain. J. Pharmacol. Exp. Ther. 358, 371–386 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Wu, Q. et al. Selective inhibitors for JNK signalling: a potential targeted therapy in cancer. J. Enzyme Inhib. Med. Chem. 35, 574–583 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Messoussi, A. et al. Recent progress in the design, study, and development of c-Jun N-terminal kinase inhibitors as anticancer agents. Chem. Biol. 21, 1433–1443 (2014).

    Article  CAS  PubMed  Google Scholar 

  228. Liu, X. & Winey, M. The MPS1 family of protein kinases. Annu. Rev. Biochem. 81, 561–585 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Schulze, V. K. et al. Treating cancer by spindle assembly checkpoint abrogation: discovery of two clinical candidates, BAY 1161909 and BAY 1217389, targeting MPS1 kinase. J. Med. Chem. 63, 8025–8042 (2020).

    Article  CAS  PubMed  Google Scholar 

  230. Lorusso, P. et al. First-in-human study of the monopolar spindle 1 (Mps1) kinase inhibitor BAY 1161909 in combination with paclitaxel in subjects with advanced malignancies. Ann. Oncol. 29, viii138 (2018).

    Article  Google Scholar 

  231. Pauklin, S., Kristjuhan, A., Maimets, T. & Jaks, V. ARF and ATM/ATR cooperate in p53-mediated apoptosis upon oncogenic stress. Biochem. Biophys. Res. Commun. 334, 386–394 (2005).

    Article  CAS  PubMed  Google Scholar 

  232. Sheils, T. K. et al. TCRD and Pharos 2021: mining the human proteome for disease biology. Nucleic Acids Res. 49, D1334–D1346 (2021).

    Article  PubMed  Google Scholar 

  233. Oprea, T. I. et al. Unexplored therapeutic opportunities in the human genome. Nat. Rev. Drug Discov. 17, 317–332 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Zawistowski, J. S. et al. Enhancer remodeling during adaptive bypass to MEK inhibition is attenuated by pharmacologic targeting of the P-TEFb complex. Cancer Discov. 7, 302–321 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Zheng, J. et al. 2.2 A refined crystal structure of the catalytic subunit of cAMP-dependent protein kinase complexed with MnATP and a peptide inhibitor. Acta Crystallogr. D. Biol. Crystallogr. 49, 362–365 (1993).

    Article  CAS  PubMed  Google Scholar 

  236. Rask-Andersen, M., Almén, M. S. & Schiöth, H. B. Trends in the exploitation of novel drug targets. Nat. Rev. Drug Discov. 10, 579–590 (2011).

    Article  CAS  PubMed  Google Scholar 

  237. Rask-Andersen, M., Masuram, S. & Schiöth, H. B. The druggable genome: evaluation of drug targets in clinical trials suggests major shifts in molecular class and indication. Annu. Rev. Pharmacol. Toxicol. 54, 9–26 (2014).

    Article  CAS  PubMed  Google Scholar 

  238. Attwood, M. M., Rask-Andersen, M. & Schiöth, H. B. Orphan drugs and their impact on pharmaceutical development. Trends Pharmacol. Sci. 39, 525–535 (2018).

    Article  CAS  PubMed  Google Scholar 

  239. Kannaiyan, R. & Mahadevan, D. A comprehensive review of protein kinase inhibitors for cancer therapy. Expert Rev. Anticancer. Ther. 18, 1249–1270 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Ferguson, F. M. & Gray, N. S. Kinase inhibitors: the road ahead. Nat. Rev. Drug Discov. 17, 353–377 (2018).

    Article  CAS  PubMed  Google Scholar 

  241. Ochoa, D. et al. Open Targets Platform: supporting systematic drug–target identification and prioritisation. Nucleic Acids Res. 49, D1302–D1310 (2021).

    Article  PubMed  Google Scholar 

  242. Wishart, D. S. et al. DrugBank: a comprehensive resource for in silico drug discovery and exploration. Nucleic Acids Res. 34, D668–D672 (2006).

    Article  CAS  PubMed  Google Scholar 

  243. 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).

    Article  CAS  PubMed  Google Scholar 

  244. Gray, N. S. & Fabbro, D. Discovery of allosteric Bcr–Abl inhibitors from phenotypic screen to clinical candidate. Methods Enzymol. 548, 173–188 (2014).

    Article  CAS  PubMed  Google Scholar 

  245. Fabbro, D. et al. Inhibitors of the Abl kinase directed at either the ATP- or myristate-binding site. Biochim. Biophys. Acta 1804, 454–462 (2010).

    Article  CAS  PubMed  Google Scholar 

  246. Hantschel, O., Rix, U. & Superti-Furga, G. Target spectrum of the BCR-ABL inhibitors imatinib, nilotinib and dasatinib. Leuk. Lymphoma 49, 615–619 (2008).

    Article  CAS  PubMed  Google Scholar 

  247. Manley, P. W. et al. Extended kinase profile and properties of the protein kinase inhibitor nilotinib. Biochim. Biophys. Acta 1804, 445–453 (2010).

    Article  CAS  PubMed  Google Scholar 

  248. Weisberg, E. et al. AMN107 (nilotinib): a novel and selective inhibitor of BCR-ABL. Br. J. Cancer 94, 1765–1769 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Huang, W. S. et al. Discovery of 3-[2-(imidazo[1,2-b]pyridazin-3-yl)ethynyl]-4-methyl-N-{4-[(4-methylpiperazin-1-yl)methyl]-3-(trifluoromethyl)phenyl}benzamide (AP24534), a potent, orally active pan-inhibitor of breakpoint cluster region-Abelson (BCR-ABL) kinase including the T315I gatekeeper mutant. J. Med. Chem. 53, 4701–4719 (2010).

    Article  CAS  PubMed  Google Scholar 

  250. Adrián, F. J. et al. Allosteric inhibitors of Bcr-abl–dependent cell proliferation. Nat. Chem. Biol. 2, 95–102 (2006).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  252. Schoepfer, J. et al. Discovery of asciminib (ABL001), an allosteric inhibitor of the tyrosine kinase activity of BCR-ABL1. J. Med. Chem. 61, 8120–8135 (2018).

    Article  CAS  PubMed  Google Scholar 

  253. Mauro, M. J. et al. A multicenter, randomized phase III study of asciminib (ABL001) versus bosutinib in patients (pts) with chronic myeloid leukemia in chronic phase (CML-CP) previously treated with ≥2 tyrosine kinase inhibitors (TKIs). J. Clin. Oncol. 37, TPS7070–TPS7070 (2019).

    Article  Google Scholar 

  254. Harris, P. A. et al. DNA-encoded library screening identifies benzo[b][1,4]oxazepin-4-ones as highly potent and monoselective receptor interacting protein 1 kinase inhibitors. J. Med. Chem. 59, 2163–2178 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

H.B.S. was supported by the Swedish Research Council, the Novo Nordisk Foundation and the Swedish Cancer Foundation. M.M.A. received support from the E. and O. Börjesons Foundation. S.K. acknowledges funding by the SGC, a registered charity that receives funds from AbbVie, Bayer, Boehringer Ingelheim, Canada Foundation for Innovation, Eshelman Institute for Innovation, Genentech, Genome Canada through Ontario Genomics Institute (OGI-196), EU/EFPIA/OICR/McGill/KTH/Diamond, Innovative Medicines Initiative 2 Joint Undertaking [EUbOPEN grant 875510], Janssen, Merck KGaA (also known as EMD in Canada and the US), Merck & Co. (also known as MSD outside Canada and the USA), Pfizer, São Paulo Research Foundation-FAPESP, Takeda and Wellcome and the Frankfurt Cancer Institute, as well as the German translational cancer network (DKTK). The authors would like to thank E. Faccenda of IUPHAR Guide to Pharmacology for her invaluable help with curating the kinase inhibitors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Helgi B. Schiöth.

Ethics declarations

Competing interests

D.F. is employed by Cellestia Biotech. Cellestia Biotech had no role in the design of the study; in the collection, analyses or interpretation of data; in the writing of the manuscript or in the decision to publish the results. The other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Drug Discovery thanks R. Roskoski, B. Murray and C. Asquith for their contribution to the peer review of this work.

Publisher’s note

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

Related links

AB Science press release: AB Science communicates the results from the phase 2B/3 study evaluating masitinib in Alzheimer disease: https://www.ab-science.com/results-from-phase-2b-3-study-evaluating-masitinib-in-alzheimers-disease/

Database of FDA-approved of drugs: https://www.accessdata.fda.gov/scripts/cder/daf/

Denali press release: Denali Therapeutics announces significant program progress and expected key milestones in 2021 for its broad therapeutic portfolio in neurodegeneration: https://www.denalitherapeutics.com/investors/press-release?id=7881&type=api

EIP Pharma neflamapimod’s clinical results release: https://www.eippharma.com/clinical-results/

Gilead press release: Gilead Receives Complete Response Letter for Filgotinib for the Treatment of Moderately to Severely Active Rheumatoid Arthritis: https://investors.gilead.com/news-releases/news-release-details/gilead-receives-complete-response-letter-filgotinib-treatment

IDG and Nature Reviews Drug Discovery Target Watch: https://commonfund.nih.gov/IDG/NRDD

Illuminating the Druggable Genome (IDG) portal: https://druggablegenome.net/

Information regarding clinical trials registered through the U.S.: https://clinicaltrials.gov/

KLIFS: https://klifs.net/

Lexicon press release: Lexicon Pharmaceuticals receives fast track designation from the FDA for LX9211 for diabetic peripheral neuropathic pain: https://www.lexpharma.com/media-Centre/news/768-lexicon-pharmaceuticals-receives-fast-track-designation-from-the-fda-for-lx9211-for-diabetic-peripheral-neuropathic-pain

Pfizer press release: Pfizer receives breakthrough therapy designation from FDA for PF-06651600, an oral JAK3 inhibitor, for the treatment of patients with alopecia areata: https://www.pfizer.com/news/press-release/press-release-detail/pfizer_receives_breakthrough_therapy_designation_from_fda_for_pf_06651600_an_oral_jak3_inhibitor_for_the_treatment_of_patients_with_alopecia_areata

Pharos interface to the Knowledge Management Center for the IDG: https://pharos.nih.gov/

Protein Data Bank (PDB): https://www.rcsb.org/

Resources for tool compounds: https://www.ebi.ac.uk/chembl/, https://pubchem.ncbi.nlm.nih.gov/, https://www.guidetopharmacology.org/, https://www.chemicalprobes.org/

Structural Genomics Consortium (SGC): https://www.thesgc.org/

Weekly updates of clinical developments of drugs: https://www.centerwatch.com/

Supplementary information

Glossary

Eukaryotic protein kinases

(ePKs). There are eight main groups of eukaryotic protein kinases: AGC (protein kinase A, G and C); CaMK (calcium/calmodulin-dependent kinases); CMGC (cyclin-dependent kinases, MAP kinases, glycogen synthase kinases, CDC-like kinases); TK (tyrosine kinases); STE (homologues of sterile 7); CK1 (casein kinases); TKL (tyrosine kinase like); and the RGC (receptor guanylate cyclase) groups.

Molecular glues

In the context of targeted protein degradation, molecular glues are small molecules that induce association of ubiquitin ligases with their target via monovalent interactions. However, designing a small molecule that can bind to both a ubiquitin ligase and a kinase in this way is highly challenging, and so molecular glues are most often identified by screening compound libraries.

Proteolysis-targeting chimeras

(PROTACs). Bivalent macromolecules composed of a flexible linker that is capped with protein-binding moieties designed to bring ubiquitin ligases and a target such as the kinase of interest into close proximity to promote its degradation. Although this strategy could enable more specific targeting of a given kinase, the large molecular size of PROTACs may pose challenges related to drug characteristics such as oral bioavailability.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Attwood, M.M., Fabbro, D., Sokolov, A.V. et al. Trends in kinase drug discovery: targets, indications and inhibitor design. Nat Rev Drug Discov 20, 839–861 (2021). https://doi.org/10.1038/s41573-021-00252-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-021-00252-y

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research