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

New kinase and HDAC hybrid inhibitors: recent advances and perspectives

    Karoline Waitman

    Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil

    &
    Roberto Parise-Filho

    *Author for correspondence: Tel.: +55 113 091 3793;

    E-mail Address: roberto.parise@usp.br

    Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil

    Published Online:https://doi.org/10.4155/fmc-2021-0276

    Cancer is the second most common cause of death worldwide. It can easily acquire resistance to treatments, demanding new therapeutic strategies, such as simultaneous inhibition of kinase and HDAC enzymes with hybrid inhibitors. Different approaches to this have varied according to their targets, with a few common trends, such as the usage of heterocycle scaffolds for kinase interaction, especially pyrimidine and quinazolines, and hydroxamic acids and benzamides for HDAC inhibition. Besides the hybrid compounds developed focusing on the inhibition tyrosine kinase and receptor tyrosine kinase, many advances have occurred in the development of serine-threonine kinase/HDAC and lipid kinase/HDAC novel compounds. Here, the latest strategies employed in this research area will be reviewed, alongside trends in inhibitor design, and observed gaps will be punctuated.

    Graphical abstract

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

    References

    • 1. World Health Organization. Cancer [Internet] (2021). www.who.int/news-room/fact-sheets/detail/cancer
    • 2. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 144(5), 646–674 (2011).
    • 3. Anighoro A, Bajorath J, Rastelli G. Polypharmacology: challenges and opportunities in drug discovery. J. Med. Chem. 57(19), 7874–7887 (2014). •• Overview of polypharmacological approaches to drug design and the advantages of hybrid inhibitor usage in comparison with combined therapy.
    • 4. Jacobs JM, Ream ME, Pensak N et al. Patient experiences with oral chemotherapy: adherence, symptoms, and quality of life. JNCCN J. Natl Compr. Cancer Netw. 17(3), 221–228 (2019).
    • 5. Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat. Rev. Cancer. 9(1), 28–39 (2009).
    • 6. Grant SK. Therapeutic protein kinase inhibitors. Cell. Mol. Life Sci. 66(7), 1163–1177 (2009).
    • 7. Ferguson FM, Gray NS. Kinase inhibitors: the road ahead. Nat. Rev. Drug Discov. 17(5), 353–376 (2018).
    • 8. Knight ZA, Lin H, Shokat KM. Targeting the cancer kinome through polypharmacology. Nat. Rev. Cancer 10, 130–137 (2010).
    • 9. Hesham HM, Lasheen DS, Abouzid KAM. Chimeric HDAC inhibitors: comprehensive review on the HDAC-based strategies developed to combat cancer. Med. Res. Rev. 38(6), 2058–2109 (2018).
    • 10. Timp W, Feinberg AP. Cancer as a dysregulated epigenome allowing cellular growth advantage at the expense of the host. Nat. Rev. Cancer 13(7), 497–510 (2013).
    • 11. Lernoux M, Schnekenburger M, Dicato M, Diederich M. Epigenetic mechanisms underlying the therapeutic effects of HDAC inhibitors in chronic myeloid leukemia. Biochem. Pharmacol. 173, 113698 (2020).
    • 12. Fortin S, Bérubé G. Advances in the development of hybrid anticancer drugs. Expert Opin. Drug Discov. 8(8), 1029–1047 (2013).
    • 13. Kumar SHM, Herrmann L, Tsogoeva SB. Structural hybridization as a facile approach to new drug candidates. Bioorganic Med. Chem. Lett. 30(23), 127514 (2020).
    • 14. Peng FW, Wu TT, Ren ZW, Xue JY, Shi L. Hybrids from 4-anilinoquinazoline and hydroxamic acid as dual inhibitors of vascular endothelial growth factor receptor-2 and histone deacetylase. Bioorganic Med. Chem. Lett. 25(22), 5137–5141 (2015).
    • 15. Peng FW, Xuan J, Wu TT et al. Design, synthesis and biological evaluation of N-phenylquinazolin-4-amine hybrids as dual inhibitors of VEGFR-2 and HDAC. Eur. J. Med. Chem. 109(2016), 1–12 (2016).
    • 16. Cai X, Zhai H-X, Wang J et al. Discovery of 7-(4-(3-Ethynylphenylamino)-7-methoxyquinazolin-6-yloxy)-N-hydroxyheptanamide (CUDC-101) as a potent multi-acting HDAC, EGFR, and HER2 inhibitor for the treatment of cancer. J. Med. Chem. 53(5), 2000–2009 (2010). •• This article describes the development of one of the most successful hybrid EGFR/HER2/HDAC inhibitors, CUDC-101, that reached clinical trials.
    • 17. Lai CJ, Bao R, Tao X et al. CUDC-101, a multitargeted inhibitor of histone deacetylase, epidermal growth factor receptor, and human epidermal growth factor receptor 2, exerts potent anticancer activity. Cancer Res. 70(9), 3647–3656 (2010).
    • 18. USA NL of M. ClinicalTrials.gov [Internet]. https://clinicaltrials.gov/
    • 19. Shimizu T, LoRusso PM, Papadopoulos KP et al. Phase I first-in-human study of CUDC-101, a multitargeted inhibitor of HDACs, EGFR, and HER2 in patients with advanced solid tumors. Clin. Cancer Res. 20(19), 5032–5040 (2014).
    • 20. Galloway TJ, Wirth LJ, Colevas AD et al. A phase I study of CUDC-101, a multitarget inhibitor of HDACs, EGFR, and HER2, in combination with chemoradiation in patients with head and neck squamous cell carcinoma. Clin. Cancer Res. 21(7), 1566–1573 (2015).
    • 21. Beckers T, Mahboobi S, Sellmer A et al. Chimerically designed HDAC- and tyrosine kinase inhibitors. A series of erlotinib hybrids as dual-selective inhibitors of EGFR, HER2 and histone deacetylases. MedChemComm 3(7), 829–835 (2012).
    • 22. Zhang X, Su M, Chen Y, Li J, Lu W. The design and synthesis of a new class of RTK/HDAC dual-targeted inhibitors. Molecules 18(6), 6491–6503 (2013).
    • 23. Ding C, Chen S, Zhang C et al. Synthesis and investigation of novel 6-(1,2,3-triazol-4-yl)-4-aminoquinazolin derivatives possessing hydroxamic acid moiety for cancer therapy. Bioorganic Med. Chem. 25(1), 27–37 (2017).
    • 24. Mahboobi S, Sellmer A, Winkler M et al. Novel chimeric histone deacetylase inhibitors: a series of lapatinib hybrides as potent inhibitors of epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and histone deacetylase activity. J. Med. Chem. 53(24), 8546–8555 (2010).
    • 25. Zuo M, Zheng YW, Lu SM, Li Y, Zhang SQ. Synthesis and biological evaluation of N-aryl salicylamides with a hydroxamic acid moiety at 5-position as novel HDAC-EGFR dual inhibitors. Bioorganic Med. Chem. 20(14), 4405–4412 (2012).
    • 26. Dong H, Yin H, Zhao C, Cao J, Xu W, Zhang Y. Design, synthesis and biological evaluation of novel osimertinib-based HDAC and EGFR dual inhibitors. Molecules 24(13), 1–17 (2019).
    • 27. Zang J, Liang X, Huang Y et al. Discovery of novel pazopanib-based HDAC and VEGFR dual inhibitors targeting cancer epigenetics and angiogenesis simultaneously. J. Med. Chem. 61(12), 5304–5322 (2018).
    • 28. Jiao Q, Bi L, Ren Y, Song S, Wang Q, Wang YS. Advances in studies of tyrosine kinase inhibitors and their acquired resistance. Mol. Cancer 17(1), 1–12 (2018).
    • 29. Lu D, Yan J, Wang L et al. Design, synthesis, and biological evaluation of the first c-Met/HDAC inhibitors based on pyridazinone derivatives. ACS Med. Chem. Lett. 8(8), 830–834 (2017).
    • 30. Xing W, Ai J, Jin S et al. Enhancing the cellular anti-proliferation activity of pyridazinones as c-Met inhibitors using docking analysis. Eur. J. Med. Chem. 95, 302–312 (2015).
    • 31. Hu H, Chen F, Dong Y, Liu Y, Gong P. Discovery of novel dual c-Met/HDAC inhibitors as a promising strategy for cancer therapy. Bioorg. Chem. 101, 103970 (2020).
    • 32. Dong Y, Hu H, Sun Y et al. Design, synthesis and biological evaluation of novel c-Met/HDAC dual inhibitors. Bioorganic Med. Chem. Lett. 30(23), 127610 (2020).
    • 33. Liu J, Qian C, Zhu Y et al. Design, synthesis and evaluate of novel dual FGFR1 and HDAC inhibitors bearing an indazole scaffold. Bioorganic Med. Chem. 26(3), 747–757 (2018).
    • 34. Liu J, Peng X, Dai Y et al. Design, synthesis and biological evaluation of novel FGFR inhibitors bearing an indazole scaffold. Org. Biomol. Chem. 13(28), 7643–7654 (2015).
    • 35. Emmons MF, Faião-Flores F, Sharma R et al. HDAC8 regulates a stress response pathway in melanoma to mediate escape from BRAF inhibitor therapy. Cancer Res. 79(11), 2947–2961 (2019).
    • 36. Zhu Y, Ran T, Chen X et al. Synthesis and biological evaluation of 1-(2-aminophenyl)-3-arylurea derivatives as potential EphA2 and HDAC dual inhibitors. Chem. Pharm. Bull. 64(8), 1136–1141 (2016).
    • 37. Chen X, Zhao S, Wu Y, Chen Y, Lu T, Zhu Y. Design, synthesis and biological evaluation of 2-amino-N-(2-aminophenyl)thiazole-5-carboxamide derivatives as novel Bcr-Abl and histone deacetylase dual inhibitors. RSC Adv. 6(105), 103178–103184 (2016).
    • 38. Liang X, Liu H, Zhang Y. Novel-targeted therapy for hematological malignancies with JAK and HDAC dual inhibitors. Future Med. Chem. 11(16), 1849–1852 (2019). • This article exemplifies how dual JAK/histone deacetylase inhibitors present reinforcing effects in cancer hallmarks, presenting a promising aproach for hematological cancer treatment.
    • 39. West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J. Clin. Invest. 124(1), 30–39 (2014).
    • 40. Chu-Farseeva YY, Mustafa N, Poulsen A et al. Design and synthesis of potent dual inhibitors of JAK2 and HDAC based on fusing the pharmacophores of XL019 and vorinostat. Eur. J. Med. Chem. 158, 593–619 (2018).
    • 41. Yang EG, Mustafa N, Tan EC et al. Design and synthesis of Janus kinase 2 (JAK2) and histone deacetlyase (HDAC) bispecific inhibitors based on pacritinib and evidence of dual pathway inhibition in hematological cell lines. J. Med. Chem. 59(18), 8233–8262 (2016).
    • 42. Liang X, Zang J, Li X et al. Discovery of novel Janus kinase (JAK) and histone deacetylase (HDAC) dual inhibitors for the treatment of hematological malignancies. J. Med. Chem. 62(8), 3898–3923 (2019).
    • 43. Liang X, Zang J, Zhu M et al. Design, synthesis, and antitumor evaluation of 4-amino-(1h)-pyrazole derivatives as JAKs inhibitors. ACS Med. Chem. Lett. 7(10), 950–955 (2016).
    • 44. Pagano L, Caira M, Candoni A et al. The epidemiology of fungal infections in patients with hematologic malignancies: the SEIFEM-2004 study. Haematologica 91(8), 1068–1075 (2006).
    • 45. Huang Y, Dong G, Li H, Liu N, Zhang W, Sheng C. Discovery of Janus kinase 2 (JAK2) and histone deacetylase (HDAC) dual inhibitors as a novel strategy for the combinational treatment of leukemia and invasive fungal infections. J. Med. Chem. 61(14), 6056–6074 (2018).
    • 46. Yao L, Mustafa N, Tan EC et al. Design and synthesis of ligand efficient dual inhibitors of Janus kinase (JAK) and histone deacetylase (HDAC) based on ruxolitinib and vorinostat. J. Med. Chem. 60(20), 8336–8357 (2017).
    • 47. Martellucci S, Clementi L, Sabetta S, Mattei V, Botta L, Angelucci A. Src family kinases as therapeutic targets in advanced solid tumors: what we have learned so far. Cancers (Basel) 12(1448), 1–28 (2020).
    • 48. Brandvold KR, Steffey ME, Fox CC, Soellner MB. Development of a highly selective c-Src kinase inhibitor. ACS Chem. Biol. 7(8), 1393–1398 (2012).
    • 49. Ko KS, Steffey ME, Brandvold KR, Soellner MB. Development of a chimeric c-Src kinase and HDAC inhibitor. ACS Med. Chem. Lett. 4(8), 779–783 (2013).
    • 50. Masliah-Planchon J, Garinet S, Pasmant E. RAS-MAPK pathway epigenetic activation in cancer: miRNAs in action. Oncotarget 7(25), 38892–38907 (2016).
    • 51. Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat. Rev. Cance. 3(6), 459–465 (2003).
    • 52. Geng A, Cui H, Zhang L et al. Discovery of novel phenoxybenzamide analogues as Raf/HDAC dual inhibitors. Bioorganic Med. Chem. Lett. 29(13), 1605–1608 (2019).
    • 53. Chen X, Gong G, Chen X et al. Design, synthesis and biological evaluation of novel benzoylimidazole derivatives as Raf and histone deacetylases dual inhibitors. Chem. Pharm. Bull. 67(10), 1116–1122 (2019).
    • 54. Jiao Y, Xin BT, Zhang Y et al. Design, synthesis and evaluation of novel 2-(1H-imidazol-2-yl) pyridine sorafenib derivatives as potential BRAF inhibitors and anti-tumor agents. Eur. J. Med. Chem. 90, 170–183 (2015).
    • 55. Cheng C, Yun F, Ullah S, Yuan Q. Discovery of novel cyclin-dependent kinase (CDK) and histone deacetylase (HDAC) dual inhibitors with potent in vitro and in vivo anticancer activity. Eur. J. Med. Chem. 189, 112073 (2020).
    • 56. Wyatt PG, Woodhead AJ Berdini V et al. Identification of N-(4-piperidinyl)-4-(2,6-dichlorobenzoylamino)-1H-pyrazole-3-carboxamide (AT7519), a novel cyclin dependent kinase inhibitor using fragment-based x-ray crystallography and structure based drug design. J. Med. Chem 51, 4986–4999 (2008).
    • 57. Yu Y, Ran D, Jiang J et al. Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2. Bioorganic Med. Chem. Lett. 29(16), 2136–2140 (2019).
    • 58. Li Y, Luo X, Guo Q et al. Discovery of N 1-(4-((7-cyclopentyl-6-(dimethylcarbamoyl)-7 H -pyrrolo[2,3- d] pyrimidin-2-yl)amino)phenyl)- N 8-hydroxyoctanediamide as a novel inhibitor targeting cyclin-dependent kinase 4/9 (CDK4/9) and histone deacetlyase1 (HDAC1) against malignant cancer. J. Med. Chem. 61(7), 3166–3192 (2018).
    • 59. D'Amore C, Borgo C, Sarno S, Salvi M. Role of CK2 inhibitor CX-4945 in anti-cancer combination therapy – potential clinical relevance. Cell. Oncol. 43(6), 1003–1016 (2020).
    • 60. Tsai SC, Seto E. Regulation of histone deacetylase 2 by protein kinase CK2. J. Biol. Chem. 277(35), 31826–31833 (2002).
    • 61. Purwin M, Hernández-Toribio J, Coderch C et al. Design and synthesis of novel dual-target agents for HDAC1 and CK2 inhibition. RSC Adv. 6(71), 66595–66608 (2016).
    • 62. Martínez R, Di Geronimo B, Pastor M et al. Multitarget anticancer agents based on histone deacetylase and protein kinase CK2 inhibitors. Molecules 25(7), 1497 (2020).
    • 63. Huang Z, Zhou W, Li Y et al. Novel hybrid molecule overcomes the limited response of solid tumours to HDAC inhibitors via suppressing JAK1-STAT3-BCL2 signalling. Theranostics 8(18), 4995–5011 (2018). • The serine-threonine kinase/tyrosine kinase/histone deacetylase inhibitor described here presented not only improved selectivity in comparison with the parental compounds but also impressive oral bioavailability, stability and capacity for tumor suppression, restricting cancer compensatory mechanisms in vivo.
    • 64. Janku F, Yap TA, Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat. Rev. Clin. Oncol. 15(5), 273–291 (2018).
    • 65. Chen Y, Yuan X, Zhang W et al. Discovery of novel dual histone deacetylase and mammalian target of rapamycin target inhibitors as a promising strategy for cancer therapy. J. Med. Chem. 62(3), 1577–1592 (2019).
    • 66. Nowak P, Cole DC, Brooijmans N et al. Discovery of potent and selective inhibitors of the mammalian target of rapamycin (mTOR) kinase. J. Med. Chem. 52(22), 7081–7089 (2009).
    • 67. Millis SZ, Ikeda S, Reddy S, Gatalica Z, Kurzrock R. Landscape of phosphatidylinositol-3-kinase pathway alterations across 19 784 diverse solid tumors. JAMA Oncol. 2(12), 1565–1573 (2016).
    • 68. Qian C, Lai CJ, Bao R et al. Cancer network disruption by a single molecule inhibitor targeting both histone deacetylase activity and phosphatidylinositol 3-kinase signaling. Clin. Cancer Res. 18(15), 4104–4113 (2012). •• This article describes the development of CUDC-907, a hybrid PI3K/histone deacetylase inhibitor with a promising profile that was able to achieve clinical trials.
    • 69. Younes A, Berdeja JG, Patel MR et al. Phase 1 safety and dose escalation of CUDC-907, a first-in-class, oral, dual inhibitor of HDAC and PI3K in relapsed or refractory lymphoma and multiple myeloma. Lancet Oncol. 17(5), 622–631 (2016).
    • 70. Oki Y, Kelly KR, Flinn I et al. CUDC-907 in relapsed/refractory diffuse large B-cell lymphoma, including patients with MYC-alterations: results from an expanded phase I trial. Haematologica 102(11), 1923–1930 (2017).
    • 71. Landsburg DJ, Barta SK, Ramchandren R et al. Fimepinostat (CUDC-907) in patients with relapsed/refractory diffuse large B cell and high-grade B-cell lymphoma: report of a phase 2 trial and exploratory biomarker analyses. Br. J. Haematol. 195(2), 201–209 (2021).
    • 72. Chen Y, Wang X, Xiang W et al. Development of purine-based hydroxamic acid derivatives: potent histone deacetylase inhibitors with marked in vitro and in vivo antitumor activities. J. Med. Chem. 59(11), 5488–5504 (2016).
    • 73. Chen D, Soh CK, Goh WH, Wang H. Design, synthesis, and preclinical evaluation of fused pyrimidine-based hydroxamates for the treatment of hepatocellular carcinoma. J. Med. Chem. 61(4), 1552–1575 (2018).
    • 74. Rodrigues DA, Guerra FS, Sagrillo FS et al. Design, synthesis, and pharmacological evaluation of first-in-class multitarget N-acylhydrazone derivatives as selective HDAC6/8 and PI3Kα inhibitors. ChemMedChem 15(6), 539–551 (2020).
    • 75. Rodrigues DA, Ferreira-Silva G, Ferreira AC et al. Design, synthesis and pharmacological evaluation of novel N-acylhydrazone derivatives as potent histone deacetylase 6 / 8 dual inhibitors. J. Med. Chem. 59(2), 655–670 (2015).
    • 76. Zhang K, Lai F, Lin S et al. Design, synthesis, and biological evaluation of 4-methyl quinazoline derivatives as anticancer agents simultaneously targeting phosphoinositide 3-kinases and histone deacetylases. J. Med. Chem. 62(15), 6992–7014 (2019).
    • 77. Lin S, Wang C, Ji M et al. Discovery and optimization of 2-amino-4-methylquinazoline derivatives as highly potent phosphatidylinositol 3-kinase inhibitors for cancer treatment. J. Med. Chem. 61(14), 6087–6109 (2018).
    • 78. Thakur A, Tawa GJ, Henderson MJ et al. Design, synthesis, and biological evaluation of quinazolin-4-one-based hydroxamic acids as dual PI3K/HDAC inhibitors. J. Med. Chem. 63(8), 4256–4292 (2020).
    • 79. Zhang L, Zhang J, Jiang Q, Zhang L, Song W. Zinc binding groups for histone deacetylase inhibitors. J. Enzyme Inhib. Med. Chem. 33(1), 714–721 (2018). • This article highlights the importance of exploring different ZBG in histone deacetylase inhibitors to achieve isoform selectivity.
    • 80. Tavares MT, Kozikowski AP, Shen S. Mercaptoacetamide: a promising zinc-binding group for the discovery of selective histone deacetylase 6 inhibitors. Eur. J. Med. Chem. 209, 112887 (2021).