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

Development and strategies of CDK4/6 inhibitors

    Pingping Chen

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China

    ,
    Yinqiu Xu

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China

    ,
    Xuanyi Li

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China

    ,
    Hequan Yao

    **Author for correspondence:

    E-mail Address: hyao@cpu.edu.cn

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China

    &
    Kejiang Lin

    *Author for correspondence:

    E-mail Address: link@cpu.edu.cn

    Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China

    Published Online:https://doi.org/10.4155/fmc-2019-0062

    Aim: CDK4/6 have critical roles in the early stage of the cell cycle. CDK2 acts later in the cell cycle and has a considerably broader range of protein substrates, some of which are essential for normal cell proliferation. Therefore, increasing the selectivity of cyclin-dependent kinase (CDK) inhibitors is critical. Methodology: In this study, we construct a versatile, specific CDK4 pharmacophore model that not only matches well with 8119 of the reported 9349 CDK4/6 inhibitors but also differentiates from the CDK2 pharmacophore. Results & Conclusion: we demonstrate the activity and selectivity determinants of CDK4/6 selective inhibitors based on the CDK4 pharmacophore model. Finally, we propose the future optimization strategy for CDK4/6 selective inhibitors, providing a theoretical basis for further research and development of CDK4/6 selective inhibitors.

    Graphical abstract

    According to our CDK4 feature model, four decisive factors (capability as a hydrogen bond donor and acceptor, hydrophobicity, electrostatic potential) are highly desirable for potent inhibitory activity and selectivity. At present, most CDK4/6 selective inhibitors have similar skeletons and branches. The structure–activity relationship revealed by the 3D quantitation structure–activity relationships pharmacophore study is illustrated in the following picture. The key hydrogen bond with the hinge region (Val96) is indispensable for activity, although it does not play a major role in selectivity. Along the dotted line, we can design a hetero atom-containing group to form a hydrogen bond interaction with the Lys35CDK4 at the bottom of the binding site. Selectivity is not the effect of a particular residue, but a cumulative effect. A unique hydrogen bonding between the side chain of His95 and the CDK4 inhibitor could improve the selectivity. There is no steric hindrance (Thr102) at the entrance of the binding pocket of CDK4, and the branch A of the scaffold could be designed to be larger such that it is appropriately positively charged and extends to the ‘top’ of the binding site (Asp99) to improve selectivity.

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

    References

    • 1. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat. Rev. Cancer 9(3), 153–166 (2009).
    • 2. Nigg EA. Cyclin-dependent protein kinases: key regulators of the eukaryotic cell cycle. BioEssays 17(6), 471–480 (1995).
    • 3. Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 140(15), 3079–3093 (2013).
    • 4. Xu Y, Chen P, Lin X, Yao H, Lin K. Discovery of CDK4 inhibitors by convolutional neural networks. Future Med. Chem. 11(3), 165–177 (2019).
    • 5. Malumbres M. Cyclin-dependent kinases. Genome Biol. 15(6), 122 (2014).
    • 6. Whittaker SR, Mallinger A, Workman P, Clarke PA. Inhibitors of cyclin-dependent kinases as cancer therapeutics. Pharmacol. Ther. 173, 83–105 (2017).
    • 7. Okamoto M, Hidaka A, Toyama M et al. Selective inhibition of HIV-1 replication by the CDK9 inhibitor FIT-039. Antiviral Res. 123, 1–4 (2015).
    • 8. Ren S, Rollins BJ. Cyclin C/cdk3 promotes Rb-dependent G0 exit. Cell 117(2), 239–251 (2004).
    • 9. Malumbres M, Barbacid M. Mammalian cyclin-dependent kinases. Trends Biochem. Sci. 30(11), 630–641 (2005).
    • 10. Dorand RD, Nthale J, Myers JT et al. Cdk5 disruption attenuates tumor PD-L1 expression and promotes antitumor immunity. Science 353(6297), 399–403 (2016).
    • 11. Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat. Rev. Drug Discov. 14(2), 130–146 (2015). •• The authors demonstrate the advantages of CDK4/6 as an antitumor target.
    • 12. Ma TL, Van Tine BA, Wei Y et al. Cell cycle-regulated phosphorylation of p220(NPAT) by cyclin E/Cdk2 in Cajal bodies promotes histone gene transcription. Genes Dev. 14(18), 2298–2313 (2000).
    • 13. Okuda M, Horn HF, Tarapore P et al. Nucleophosmin/B23 is a target of CDK2/Cyclin E in centrosome duplication. Cell 103(1), 127–140 (2000).
    • 14. Sever-Chroneos Z, Angus SP, Fribourg AF et al. Retinoblastoma tumor suppressor protein signals through inhibition of cyclin-dependent kinase 2 activity to disrupt PCNA function in S phase. Mol. Cell. Biol. 21(12), 4032–4045 (2001).
    • 15. Sherr CJ, Beach D, Shapiro GI. Targeting CDK4 and CDK6: from discovery to therapy. Cancer Discov. 6(4), 353–367 (2016). • The authors show the progress of research on CDK4/6 inhibitors.
    • 16. Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J. Clin. Oncol. 24(11), 1770–1783 (2006).
    • 17. Hamilton E, Infante JR. Targeting CDK4/6 in patients with cancer. Cancer Treat. Rev. 45, 129–138 (2016).
    • 18. Otto T, Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat. Rev. Cancer 17(2), 93–115 (2017).
    • 19. Goel S, DeCristo MJ, Watt AC et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 548(7668), 471–475 (2017). • The authors revealed that CDK4/6 inhibitors can stimulate antitumor immunity.
    • 20. Deng J, Wang ES, Jenkins RW et al. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 8(2), 216–233 (2018).
    • 21. Zhang J, Bu X, Wang H et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via Cul3(SPOP) to control cancer immune surveillance. Nature 553(7686), 91–95 (2017).
    • 22. Oh SJ, Cho H, Kim S et al. Targeting cyclin D-CDK4/6 sensitizes immune-refractory cancer by blocking the SCP3-NANOG axis. Cancer Res. 78(10), 2638–2653 (2018).
    • 23. Schaer DA, Beckmann RP, Dempsey JA et al. The CDK4/6 inhibitor abemaciclib induces a T cell inflamed tumor microenvironment and enhances the efficacy of PD-L1 checkpoint blockade. Cell. Rep. 22(11), 2978–2994 (2018).
    • 24. Mascarenhas NM, Bhattacharyya D, Ghoshal N. Why pyridine containing pyrido[2,3-d]pyrimidin-7-ones selectively inhibit CDK4 than CDK2: insights from molecular dynamics simulation. J. Mol. Graph. Model. 28(7), 695–706 (2010).
    • 25. Shafiq MI, Steinbrecher T, Schmid R. Fascaplysin as a specific inhibitor for CDK4: insights from molecular modelling. PLoS ONE 7(8), e42612 (2012). •• The author constructed a hybrid CDK4 structure.
    • 26. Barvian M, Boschelli DH, Cossrow J et al. Pyrido[2,3-d]pyrimidin-7-one inhibitors of cyclin-dependent kinases. J. Med. Chem. 43(24), 4606–4616 (2000).
    • 27. Toogood PL, Harvey PJ, Repine JT et al. Discovery of a potent and selective inhibitor of cyclin-dependent kinase 4/6. J. Med. Chem. 48(7), 2388–2406 (2005).
    • 28. VanderWel SN, Harvey PJ, McNamara DJ et al. Pyrido[2,3-d]pyrimidin-7-ones as specific inhibitors of cyclin-dependent kinase 4. J. Med. Chem. 48(7), 2371–2387 (2005).
    • 29. Reddy MV, Akula B, Cosenza SC et al. Discovery of 8-cyclopentyl-2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-7-oxo-7,8-dihydro-pyrid o[2,3-d]pyrimidine-6-carbonitrile (7x) as a potent inhibitor of cyclin-dependent kinase 4 (CDK4) and AMPK-related kinase 5 (ARK5). J. Med. Chem. 57(3), 578–599 (2014).
    • 30. Moriarty KJ, Koblish HK, Garrabrant T et al. The synthesis and SAR of 2-amino-pyrrolo[2,3-d]pyrimidines: a new class of Aurora-A kinase inhibitors. Bioorg. Med. Chem. Lett. 16(22), 5778–5783 (2006).
    • 31. Cho YS, Borland M, Brain C et al. 4-(Pyrazol-4-yl)-pyrimidines as selective inhibitors of cyclin-dependent kinase 4/6. J. Med. Chem. 53(22), 7938–7957 (2010).
    • 32. Li Z, Wang X, Eksterowicz J et al. Discovery of AMG 925, a FLT3 and CDK4 dual kinase inhibitor with preferential affinity for the activated state of FLT3. J. Med. Chem. 57(8), 3430–3449 (2014).
    • 33. Horiuchi T, Chiba J, Uoto K, Soga T. Discovery of novel thieno[2,3-d]pyrimidin-4-yl hydrazone-based inhibitors of Cyclin D1-CDK4: synthesis, biological evaluation, and structure-activity relationships. Bioorg. Med. Chem. Lett. 19(2), 305–308 (2009).
    • 34. Horiuchi T, Nagata M, Kitagawa M, Akahane K, Uoto K. Discovery of novel thieno[2,3-d]pyrimidin-4-yl hydrazone-based inhibitors of Cyclin D1-CDK4: synthesis, biological evaluation and structure–activity relationships. Part 2. Bioorg. Med. Chem. 17(23), 7850–7860 (2009).
    • 35. Cai BQ, Jin HX, Yan XJ, Zhu P, Hu GX. 3D-QSAR and 3D-QSSR studies of thieno[2,3-d]pyrimidin-4-yl hydrazone analogues as CDK4 inhibitors by CoMFA analysis. Acta Pharmacol. Sin. 35(1), 151–160 (2014). • The authors used the 3D quantitation structure–activity relationships pharmacophore model to study CDK4 inhibitors.
    • 36. Beattie JF, Breault GA, Ellston RP et al. Cyclin-dependent kinase 4 inhibitors as a treatment for cancer. Part 1: identification and optimisation of substituted 4,6-bis anilino pyrimidines. Bioorg. Med. Chem. Lett. 13(18), 2955–2960 (2003).
    • 37. Breault GA, Ellston RP, Green S et al. Cyclin-dependent kinase 4 inhibitors as a treatment for cancer. Part 2: identification and optimisation of substituted 2,4-bis anilino pyrimidines. Bioorg. Med. Chem. Lett. 13(18), 2961–2966 (2003).
    • 38. McInnes C, Wang S, Anderson S et al. Structural determinants of CDK4 inhibition and design of selective ATP competitive inhibitors. Chem. Biol. 11(4), 525–534 (2004).
    • 39. Wang SD, Meades C, Wood G et al. 2-Anilino-4-(thiazol-5-yl)pyrimidine CDK inhibitors: Synthesis, SAR analysis, X-ray crystallography, and biological activity. J. Med. Chem. 47(7), 1662–1675 (2004).
    • 40. Tadesse S, Yu M, Mekonnen LB et al. Highly potent, selective, and orally bioavailable 4-Thiazol-N-(pyridin-2-yl)pyrimidin-2-amine cyclin-dependent kinases 4 and 6 inhibitors as anticancer drug candidates: design, synthesis, and evaluation. J. Med. Chem. 60(5), 1892–1915 (2017).
    • 41. Soni R, Muller L, Furet P et al. Inhibition of cyclin-dependent kinase 4 (CDK4) by fascaplysin, a marine natural product. Biochem. Biophys. Res. Commun. 275(3), 877–884 (2000).
    • 42. Aubry C, Jenkins PR, Mahale S, Chaudhuri B, Marechal JD, Sutcliffe MJ. New fascaplysin-based CDK4-specific inhibitors: design, synthesis and biological activity. Chem. Commun. (Camb.) (15), 1696–1697 (2004).
    • 43. Aubry C, Wilson AJ, Jenkins PR et al. Design, synthesis and biological activity of new CDK4-specific inhibitors, based on fascaplysin. Org. Biomol. Chem. 4(5), 787–801 (2006).
    • 44. Garcia MD, Wilson AJ, Emmerson DP, Jenkins PR, Mahale S, Chaudhuri B. Synthesis, crystal structure and biological activity of beta-carboline based selective CDK4-cyclin D1 inhibitors. Org. Biomol. Chem. 4(24), 4478–4484 (2006).
    • 45. Mahale S, Aubry C, James Wilson A et al. CA224, a non-planar analogue of fascaplysin, inhibits Cdk4 but not Cdk2 and arrests cells at G0/G1 inhibiting pRB phosphorylation. Bioorg. Med. Chem. Lett. 16(16), 4272–4278 (2006).
    • 46. Jenkins PR, Wilson J, Emmerson D et al. Design, synthesis and biological evaluation of new tryptamine and tetrahydro-β-carboline-based selective inhibitors of CDK4. Bioorg. Med. Chem. 16(16), 7728–7739 (2008).
    • 47. Aubry C, Wilson AJ, Emmerson D et al. Fascaplysin-inspired diindolyls as selective inhibitors of CDK4/cyclin D1. Bioorg. Med. Chem. 17(16), 6073–6084 (2009).
    • 48. Mahale S, Bharate SB, Manda S et al. Biphenyl-4-carboxylic acid [2-(1H-indol-3-yl)-ethyl]-methylamide (CA224), a nonplanar analogue of fascaplysin, inhibits Cdk4 and tubulin polymerization: evaluation of in vitro and in vivo anticancer activity. J. Med. Chem. 57(22), 9658–9672 (2014).
    • 49. Honma T, Hayashi K, Aoyama T et al. Structure-based generation of a new class of potent Cdk4 inhibitors: new de novo design strategy and library design. J. Med. Chem. 44(26), 4615–4627 (2001).
    • 50. Honma T, Yoshizumi T, Hashimoto N et al. A novel approach for the development of selective Cdk4 inhibitors: library design based on locations of Cdk4 specific amino acid residues. J. Med. Chem. 44(26), 4628–4640 (2001).
    • 51. Ikuta M, Kamata K, Fukasawa K et al. Crystallographic approach to identification of cyclin-dependent kinase 4 (CDK4)-specific inhibitors by using CDK4 mimic CDK2 protein. J. Biol. Chem. 276(29), 27548–27554 (2001).
    • 52. Tsou HR, Otteng M, Tran T et al. 4-(Phenylaminomethylene)isoquinoline-1,3(2H,4H)-diones as potent and selective inhibitors of the cyclin-dependent kinase 4 (CDK4). J. Med. Chem. 51(12), 3507–3525 (2008).
    • 53. Tsou HR, Liu X, Birnberg G et al. Discovery of 4-(benzylaminomethylene)isoquinoline-1,3-(2H,4H)-diones and 4-[(pyridylmethyl)aminomethylene]isoquinoline-1,3-(2H,4H)-diones as potent and selective inhibitors of the cyclin-dependent kinase 4. J. Med. Chem. 52(8), 2289–2310 (2009).
    • 54. Zheng J, Kong H, Wilson JM et al. Insight into the interactions between novel isoquinolin-1,3-dione derivatives and cyclin-dependent kinase 4 combining QSAR and molecular docking. PLoS ONE 9(4), e93704 (2014).
    • 55. Ryu CK, Kang HY, Lee SK et al. 5-Arylamino-2-methyl-4,7-dioxobenzothiazoles as inhibitors of cyclin-dependent kinase 4 and cytotoxic agents. Bioorg. Med. Chem. Lett. 10(5), 461–464 (2000).
    • 56. Goodman J. Reaxys. J. Chem. Inf. Model. 49(12), 2897–2898 (2009).
    • 57. Park H, Yeom MS, Lee S. Loop flexibility and solvent dynamics as determinants for the selective inhibition of cyclin-dependent kinase 4: comparative molecular dynamics simulation studies of CDK2 and CDK4. ChemBioChem 5(12), 1662–1672 (2004).
    • 58. Tutone M, Almerico AM. Recent advances on CDK inhibitors: an insight by means of in silico methods. Eur. J. Med. Chem. 142, 300–315 (2017).
    • 59. Bai J, Li Y, Zhang G. Cell cycle regulation and anticancer drug discovery. Cancer Biol. Med. 14(4), 348–362 (2017).
    • 60. Wang H, Wang K, Guan Z et al. Computational study of non-catalytic T-loop pocket on CDK proteins for drug development. Chinese Phys. B 26(12), 128702 (2017). • The authors introduced non-ATP competitive cyclin-dependent kinase inhibitors.
    • 61. Caballero J, Fernandez M, Gonzalez-Nilo FD. Structural requirements of pyrido[2,3-d]pyrimidin-7-one as CDK4/D inhibitors: 2D autocorrelation, CoMFA and CoMSIA analyses. Bioorg. Med. Chem. 16(11), 6103–6115 (2008).
    • 62. Mascarenhas NM, Ghoshal N. Combined ligand and structure based approaches for narrowing on the essential physicochemical characteristics for CDK4 inhibition. J. Chem. Inf. Model. 48(7), 1325–1336 (2008). • The authors show the key amino acids of CDK2, CDK4 and CDK6 active sites.
    • 63. Mysinger MM, Carchia M, Irwin JJ, Shoichet BK. Directory of useful decoys, enhanced (DUD-E): better ligands and decoys for better benchmarking. J. Med. Chem. 55(14), 6582–6594 (2012). • The author introduced a decoy set.