We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Achieving effective and selective CK1 inhibitors through structure modification

    Chenxi Du

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China

    ,
    Hongyu Yang

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China

    ,
    Feng Feng

    Jiangsu Food & Pharmaceutical Science College, No.4 Meicheng Road, Huai'an 223003, People’s Republic of China

    Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People’s Republic of China

    ,
    Wenyuan Liu

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China

    ,
    Yao Chen

    School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China

    &
    Haopeng Sun

    *Author for correspondence:

    E-mail Address: sunhaopeng@163.com

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China

    Jiangsu Food & Pharmaceutical Science College, No.4 Meicheng Road, Huai'an 223003, People’s Republic of China

    Published Online:https://doi.org/10.4155/fmc-2020-0215

    Casein kinase 1 (CK1) is an extensively expressed serine/threonine kinase family, with six highly conserved isoforms of human CK1. Due to its involvement in many biological processes, CK1 is a promising target for several pathological states, including circadian sleep disorder, neurodegenerative diseases, cancer and inflammation. However, due to the structural similarities between the six CK1 members, the design of CK1 inhibitors is intricate. So far, no effective CK1 inhibitors are reported to reach clinical trials; thus, approaches to obtaining both selective and effective CK1 inhibitors are in great demand. Here we analyze several CK1 inhibitors that provide successful experience for structure-based drug design and rational structure modification, which could provide references for further drug design.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Rowles J, Slaughter C, Moomaw C, Hsu J, Cobb MH. Purification of casein kinase I and isolation of cDNAs encoding multiple casein kinase I-like enzymes. Proc. Natl Acad. Sci. USA 88, 9548–9552 (1991).
    • 2. Knippschild U, Gocht A, Wolff S, Huber N, Lohler J, Stoter M. The casein kinase 1 family: participation in multiple cellular processes in eukaryotes. Cell. Signal. 17(6), 675–689 (2005).
    • 3. Gross SD, Anderson RA. Casein kinase I: spatial organization and positioning of a multifunctional protein kinase family. Cell. Signal. 10(10), 699–711 (1998).
    • 4. Knippschild U, Wolff S, Giamas G et al. The role of the casein kinase 1 (CK1) family in different signaling pathways linked to cancer development. Onkologie 28(10), 508–514 (2005).
    • 5. Budini M, Jacob G, Jedlicki A, Perez C, Allende CC, Allende JE. Autophosphorylation of carboxy-terminal residues inhibits the activity of protein kinase CK1alpha. J. Cell. Biochem. 106(3), 399–408 (2009).
    • 6. Dhillon N, Hoekstra MF. Characterization of two protein kinases from Schizosaccharomyces pombe involved in the regulation of DNA repair. EMBO J. 13(12), 2777–2788 (1994).
    • 7. Lohler J, Hirner H, Schmidt B et al. Immunohistochemical characterisation of cell-type specific expression of CK1delta in various tissues of young adult BALB/c mice. PLoS ONE 4(1), 12 (2009).
    • 8. Panchenko MP, Siddiquee Z, Dombkowski DM et al. Protein kinase CK1alphaLS promotes vascular cell proliferation and intimal hyperplasia. Am. J. Pathol. 177(3), 1562–1572 (2010).
    • 9. Cheong JK, Nguyen TH, Wang H et al. IC261 induces cell cycle arrest and apoptosis of human cancer cells via CK1δ/ε and Wnt/β-catenin independent inhibition of mitotic spindle formation. Oncogene 30(22), 2558–2569 (2011).
    • 10. Meng QJ, Maywood ES, Bechtold DA et al. Entrainment of disrupted circadian behavior through inhibition of casein kinase 1 (CK1) enzymes. Proc. Natl Acad. Sci. USA 107(34), 15240–15245 (2010).
    • 11. Curtis AT, Alison JH, Judsen S et al. Small-molecule inhibition of Wnt signaling through activation of casein kinase 1alpha. Nat. Chem. Biol. 6, 829–836 (2010).
    • 12. Jiang J. CK1 in developmental signaling: Hedgehog and Wnt. Curr. Top. Dev. Biol. 123, 303 (2016).
    • 13. Niehrs C. The complex world of WNT receptor signalling. Nat. Rev. Mol. Cell Biol. 13(12), 767–779 (2012).
    • 14. Cruciat CM. Casein kinase 1 and Wnt/β-catenin signaling. Curr. Opin. Cell Biol. 31, 46–55 (2014).
    • 15. Davidson G, Wu W, Shen J et al. Casein kinase 1 gamma couples Wnt receptor activation to cytoplasmic signal transduction. Nature 438(7069), 867–872 (2005).
    • 16. Swiatek W, Kang H, Garcia BA et al. Negative regulation of LRP6 function by casein kinase Iepsilon phosphorylation. J. Biol. Chem. 281(18), 12233–12241 (2006).
    • 17. Cruciat CM, Niehrs C. Secreted and transmembrane Wnt inhibitors and activators. Cold Spring Harb. Perspect. Biol. 5(3), a015081 (2013).
    • 18. Meng QJ, Maywood ES, Bechtold DA et al. Entrainment of disrupted circadian behavior through inhibition of casein kinase 1 (CK1) enzymes. Proc. Natl Acad. Sci. USA 24(107), 15240–15245 (2010).
    • 19. Merrow M, Mazzotta G, Chen Z et al. The right place at the right time: regulation of daily timing by phosphorylation. Genes Dev. 20(19), 2629–2633 (2006).
    • 20. Camacho F, Cilio M, Guo Y et al. Human casein kinase I delta phosphorylation of human circadian clock proteins period 1 and 2. FEBS Lett. 489(2–3), 159–165 (2001).
    • 21. Lee C, Etchegaray JP, Cagampang RA et al. Posttranslational mechanisms regulate the mammalian circadian clock. Cell 107(7), 855–867 (2001).
    • 22. Vielhaber E, Eide E, Rivers A et al. Nuclear entry of the circadian regulator mPER1 is controlled by mammalian casein kinase Iε. Mol. Cell. Biol. 20(13), 4888–4899 (2000).
    • 23. Akashi M, Tsuchiya Y, Yoshino T et al. Control of intracellular dynamics of mammalian period proteins by casein kinase 1 epsilon (CK1 epsilon) and CK1delta in cultured cells. Mol. Cell. Biol. 22(6), 1693–1703 (2002).
    • 24. Price JL, Blau J, Rothenfluh A et al. Double-time is a novel Drosophila clock gene that regulates PERIOD protein accumulation. Cell 94(1), 83–95 (1998).
    • 25. Hirota T, Lee JW, Lewis WG et al. High-throughput chemical screen identifies a novel potent modulator of cellular circadian rhythms and reveals CKIα as a clock regulatory kinase. PLoS Biol. 8(12), 14 (2010).
    • 26. Li G, Yin H, Kuret J. Casein kinase 1 delta phosphorylates tau and disrupts its binding to microtubules. J. Biol. Chem. 279(16), 15938–15945 (2004).
    • 27. Xu Y, Padiath QS, Shapiro RE et al. Functional consequence of a CK1δ mutation causing familial advanced sleep phase syndrome. Nature 434, 640–644 (2005).
    • 28. Yang WS, Stockwell BR. Inhibition of casein kinase 1-epsilon induces cancer-cell-selective, PERIOD2-dependent growth arrest. Genome Biol. 9(6), R92 (2008).
    • 29. Longenecker KL, Roach PJ, Hurley TD. Three-dimensional structure of mammalian casein kinase I: molecular basis for phosphate recognition. J. Mol. Biol. 257(3), 618–631 (1996).
    • 30. Knippschild U, Kruger M, Richter J et al. The CK1 family: contribution to cellular stress response and its role in carcinogenesis. Front. Oncol. 4, 96 (2014).
    • 31. Xu RM, Carmel G, Sweet RM et al. Crystal structure of casein kinase-1, a phosphate-directed protein kinase. EMBO J. 14(5), 1015–1023 (1995).
    • 32. Luxenburger A, Schmidt D, Ianes C et al. Design, synthesis and biological evaluation of isoxazole-based CK1 inhibitors modified with chiral pyrrolidine scaffolds. Molecules 24(5), 873 (2019).
    • 33. Peifer C, Abadleh M, Bischof J et al. 3,4-diaryl-isoxazoles and -imidazoles as potent dual inhibitors of p38α mitogen activated protein kinase and casein kinase 1δ. J. Med. Chem. 52(23), 7618–7630 (2009).
    • 34. Mian A, Schull M, Zhao Z et al. The gatekeeper mutation T315I confers resistance against small molecules by increasing or restoring the ABL-kinase activity accompanied by aberrant transphosphorylation of endogenous BCR, even in loss-of-function mutants of BCR/ABL. Leukemia 23(9), 1614–1621 (2009).
    • 35. Rodrigues RP, Da Silva C. Discovery of potential neurodegenerative inhibitors in Alzheimer’s disease by casein kinase 1 structure-based virtual screening. Med. Chem. Res. 26(12), 3274–3285 (2017).
    • 36. Flotow H, Graves PR, Wang AQ et al. Phosphate groups as substrate determinants for casein kinase I action. J. Biol. Chem. 265(24), 14264–14269 (1990).
    • 37. Roach PJ. Multisite and hierarchal protein phosphorylation. J. Biol. Chem. 266(22), 14139–14142 (1991).
    • 38. Marin O, Bustos VH, Cesaro L et al. A noncanonical sequence phosphorylated by casein kinase 1 in beta-catenin may play a role in casein kinase 1 targeting of important signaling proteins. Proc. Natl Acad. Sci. USA 100(18), 10193–10200 (2003).
    • 39. Kawakami F, Suzuki K, Ohtsuki K. A novel consensus phosphorylation motif in sulfatide- and cholesterol-3-sulfate-binding protein substrates for CK1 in vitro. Biol. Pharm. Bull. 31(2), 193–200 (2008).
    • 40. Gebel J, Tuppi M, Chaikuad A et al. p63 uses a switch-like mechanism to set the threshold for induction of apoptosis. Nat. Chem. Biol 16(10), 1078–1086 (2020).
    • 41. Xu RM, Carmel G, Kuret J et al. Structural basis for selectivity of the isoquinoline sulfonamide family of protein kinase inhibitors. Proc. Natl Acad. Sci. USA 93(13), 6308–6313 (1996).
    • 42. Chico LK, Van Eldik LJ, Watterson DM. Targeting protein kinases in central nervous system disorders. Nat. Rev. Drug Discov. 8(11), 892–909 (2009).
    • 43. Mashhoon N, Demaggio AJ, Tereshko V et al. Crystal structure of a conformation-selective casein kinase-1 inhibitor. J. Biol. Chem. 275(26), 20052–20060 (2000).
    • 44. Brockschmidt C, Hirner H, Huber N et al. Anti-apoptotic and growth-stimulatory functions of CK1 delta and epsilon in ductal adenocarcinoma of the pancreas are inhibited by IC261 in vitro and in vivo. Gut 57(6), 799–806 (2008).
    • 45. Fohr KJ, Knippschild U, Herkommer A et al. State-dependent block of voltage-gated sodium channels by the casein-kinase 1 inhibitor IC261. Invest. New Drugs 35(3), 277–289 (2017).
    • 46. Rena G, Bain J, Elliott M, Cohen P. D4476, a cell-permeant inhibitor of CK1, suppresses the site-specific phosphorylation and nuclear exclusion of FOXO1a. EMBO Rep. 5(1), 60–65 (2004).
    • 47. Bain J, Plater L, Elliott M et al. The selectivity of protein kinase inhibitors: a further update. Biochem. J. 408(3), 297–315 (2007).
    • 48. Hu Y, Song W, Cirstea D et al. CSNK1α1 mediates malignant plasma cell survival. Leukemia 29(2), 474–482 (2014).
    • 49. Badura L, Swanson T, Adamowicz W et al. An inhibitor of casein kinase I epsilon induces phase delays in circadian rhythms under free-running and entrained conditions. J. Pharmacol. Exp. Ther. 322(2), 730–738 (2007). • In this article, the researchers discovered a highly potent and selective CK1ε inhibitor with IC50 value being 7.7 nM. Intensive pharmacology studies were performed to verify the effectiveness. In the free-running animals test, the inhibitor was demonstrated to produce significant phase delay dosing at circadian time.
    • 50. Keenan CR, Langenbach SY, Jativa F et al. Casein kinase 1δ/ε inhibitor, PF670462 attenuates the fibrogenic effects of transforming growth factor-β in pulmonary fibrosis. Front. Pharmacol. 9, 738 (2018).
    • 51. Long A, Zhao H, Huang X. Structural basis for the interaction between casein kinase 1 delta and a potent and selective inhibitor. J. Med. Chem. 55(2), 956–960 (2012).
    • 52. Walton KM, Fisher K, Rubitski D et al. Selective inhibition of casein kinase 1 epsilon minimally alters circadian clock period. J. Pharmacol. Exp. Ther. 330(2), 430–439 (2009).
    • 53. Long AM, Zhao H, Huang X. Structural basis for thepotent and selective inhibitionof casein kinase 1 epsilon. J. Med. Chem. 55(22), 10307–10311 (2012). •• Three crystal structures were reported in this article. Based on the in-depth analysis of the molecular mechanism of action, many insights for the design of selective CK1 inhibitors were put forward.
    • 54. Graneto MJ, Kurumbail RG, Vazquez ML et al. Synthesis, crystal structure, and activity of pyrazole-based inhibitors of p38 kinase. J. Med. Chem. 50(23), 5712–5719 (2007).
    • 55. Mente S, Arnold E, Butler T et al. Ligand-protein interactions of selective casein kinase 1δ inhibitors. J. Med. Chem. 56(17), 6819–6828 (2013).
    • 56. Wager TT, Galatsis P, Chandrasekaran RY et al. Identification and profiling of a selective and brain penetrant radioligand for in vivo target occupancy measurement of casein kinase 1 (CK1) inhibitors. ACS Chem. Neurosci. 8(9), 1995–2004 (2017).
    • 57. Halekotte J, Witt L, Ianes C et al. Optimized 4,5-diarylimidazoles as potent/selective inhibitors of protein kinase CK1δ and their structural relation to p38αMAPK. Molecules 22(4), 522 (2017).
    • 58. Schehr M, Ianes C, Weisner J et al. 2-azo-, 2-diazocine-thiazols and 2-azoimidazoles as photoswitchable kinase inhibitors: limitations and pitfalls of the photoswitchable inhibitor approach. Photochem. Photobiol. Sci. 18(6), 1398–1407 (2019).
    • 59. Lovering F, Bikker J, Humblet C. Escape from flatland: increasing saturation as an approach to improving clinical success. J. Med. Chem. 52(21), 6752–6756 (2009).
    • 60. Huart AS, Saxty B, Merritt A et al. A casein kinase 1/checkpoint kinase 1 pyrazolo-pyridine protein kinase inhibitor as novel activator of the p53 pathway. Bioorg. Med. Chem. Lett. 23(20), 5578–5585 (2013).
    • 61. Huang H, Acquaviva L, Berry V et al. Structure-based design of potent and selective CK1γ inhibitors. ACS Med. Chem. Lett. 3(12), 1059–1064 (2012). •• In the field of CK1 inhibitors, few studies were performed on selective CK1γ inhibitors. In this article, structure-based approach was adopted to design a series of highly potent and selectiveCK1γ inhibitors, which exhibit excellent enzyme and cell potency.
    • 62. Leban J, Baierl M, Mies J et al. A novel class of potent NF-κB signaling inhibitors. Bioorg. Med. Chem. Lett. 17(21), 5858–5862 (2007).
    • 63. Bischof J, Leban J, Zaja M et al. 2-Benzamido-N-(1H-benzo[d]imidazol-2-yl) thiazole-4-carboxamide derivatives as potent inhibitors of CK1δ/ε. Amino Acids 43(4), 1577–1591 (2012).
    • 64. Richter J, Bischof J, Zaja M et al. Difluoro-dioxolo-benzoimidazol-benzamides as potent inhibitors of CK1δ and ε with nanomolar inhibitory activity on cancer cell proliferation. J. Med. Chem. 57(19), 7933–7946 (2014). • By analyzing the structure–activity relationship, the researchers designed a series of derivatives with increased biological activity. The preferred compound is inhibitory active and available within the cell.
    • 65. Madoux F, Simanski S, Chase P et al. An ultra-high-throughput cell-based screen for wee1 degradation inhibitors. J. Biomol. Screen. 15(8), 907–917 (2010).
    • 66. Bibian M, Rahaim RJ, Choi JY et al. Development of highly selective casein kinase 1δ/1ε (CK1δ/ε) inhibitors with potent antiproliferative properties. Bioorg. Med. Chem. Lett. 23(15), 4374–4380 (2013).
    • 67. Garcia-Reyes B, Witt L, Jansen B et al. Discovery of inhibitor of wnt production 2 (IWP-2) and related compounds as selective ATP-competitive inhibitors of casein kinase 1 (CK1) δ /ε. J. Med. Chem. 61(9), 4087–4102 (2018).
    • 68. Salado IG, Redondo M, Bello ML et al. Protein kinase CK-1 inhibitors as new potential drugs for amyotrophic lateral sclerosis. J. Med. Chem. 57(6), 2755–2772 (2014). • By the approach of chemical genetic, the researchers discovered and optimized several CK1 inhibitors. In the cell assay, these compounds were able to exhibit inhibitory effects. Thus, the molecules are hopeful tools to study the therapy of ALS.
    • 69. Lee JW, Hirota T, Peters EC et al. A small molecule modulates circadian rhythms through phosphorylation of the period protein. Angew Chem. Int. Ed. Engl. 50(45), 10608–10611 (2011).
    • 70. Etchegaray JP, Machida KK, Noton E et al. Casein kinase 1 delta regulates the pace of the mammalian circadian clock. Mol. Cell. Biol. 29(14), 3853–3866 (2009).
    • 71. Miyazaki K, Nagase T, Mesaki M et al. Phosphorylation of clock protein PER1 regulates its circadian degradation in normal human fibroblasts. Biochem. J. 380(Pt 1), 95–103 (2004).
    • 72. Lee JW, Hirota T, Ono D et al. Chemical control of mammalian circadian behavior through dual inhibition of casein kinase Iα and δ. J. Med. Chem. 62(4), 1989–1998 (2019).
    • 73. Yang LL, Li GB, Yan HX et al. Discovery of N6-phenyl-1H-pyrazolo[3,4-d]pyrimidine-3,6-diamine derivatives as novel CK1 inhibitors using common-feature pharmacophore model based virtual screening and hit-to-lead optimization. Eur. J. Med. Chem. 56, 30–38 (2012).
    • 74. Lee MR, Dominguez C. MAP kinase p38 inhibitors: clinical results and an intimate look at their interactions with p38 protein. Curr. Med. Chem. 12(25), 2979–2994 (2005).
    • 75. Miwatashi S, Arikawa Y, Kotani E et al. Novel inhibitor of p38 MAP kinase as an anti-TNF-α drug: discovery of N-[4-[2-ethyl-4-(3-methylphenyl)-1,3-thiazol-5-yl]-2-pyridyl]benzamide (TAK-715) as a potent and orally active anti-rheumatoid arthritis agent. J. Med. Chem. 48(19), 5966–5979 (2005).
    • 76. Verkaar F, Van Der Doelen AA, Smits JF, Blankesteijn WM, Zaman GJ. Inhibition of Wnt/β-catenin signaling by p38 MAP kinase inhibitors is explained by cross-reactivity with casein kinase Iδ/ε. Chem. Biol. 18(4), 485–494 (2011).
    • 77. Myrianthopoulos V, Lozach O, Zareifi D et al. Combined virtual and experimental screening for CK1 inhibitors identifies a modulator of p53 and reveals important aspects of in silico screening performance. Int. J. Mol. Sci. 18(10), 2102 (2017).
    • 78. List A, Dewald G, Bennett J et al. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N. Engl. J. Med. 355(14), 1456–1465 (2006).
    • 79. Kronke J, Fink EC, Hollenbach PW et al. Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS. Nature 523(7559), 183–188 (2015).
    • 80. Petzold G, Fischer ES, Thoma NH. Structrual basis of lenalidomide-induced CK1α degradation by the CRL4CRBN ubiquitin ligase. Nature 532(7597), 127–130 (2016). •• In the research field of CK1, protein–protein interaction are rarely mentioned. In this article, the author reported the crystal structure of DDB1–CRBN bound to lenalidomide and CK1α, providing novel ideas for future drug design.
    • 81. Yang Y, Shaffer Arthur L, Emre NCT et al. Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma. Cancer Cell 21(6), 723–737 (2012).
    • 82. Minzel W, Venkatachalam A, Flink A et al. Small molecules co-targeting CK1α and the transcriptional kinase CDK7/9 control AML in preclinical models. Cell 175(1), 171–185 (2018).
    • 83. Kojima K, Ishizawa J, Andreeff M. Pharmacological activation of wild-type p53 in the therapy of leukemia. Exp. Hematol. 44(9), 791–798 (2016).
    • 84. Klaeger S, Heinzlmeir S, Wilhelm M et al. The target landscape of clinical kinase drugs. Science 358(6367), eaan4368 (2017).