566
Views
7
CrossRef citations to date
0
Altmetric
Research Articles

Identification of HDAC6 selective inhibitors: pharmacophore based virtual screening, molecular docking and molecular dynamics simulation

, , , , , , , , & show all
Pages 1928-1939 | Received 09 Aug 2019, Accepted 03 Mar 2020, Published online: 11 Apr 2020

References

  • Alves Avelar, L. A., Ruzic, D., Djokovic, N., Kurz, T., & Nikolic, K. (2019). Structure-based design of selective histone deacetylase 6 zinc binding groups. Journal of Biomolecular Structure and Dynamics, 1–12. doi:10.1080/07391102.2019.1652687
  • Amin, S. A., Adhikari, N., & Jha, T. (2019). Development of decision trees to discriminate HDAC8 inhibitors and non-inhibitors using recursive partitioning. Journal of Biomolecular Structure and Dynamics, 1–8. doi:10.1080/07391102.2019.1661876
  • Amin, S. A., Adhikari, N., Jha, T., & Ghosh, B. (2019). Designing potential HDAC3 inhibitors to improve memory and learning. Journal of Biomolecular Structure and Dynamics, 37(8), 2133–2142. doi:10.1080/07391102.2018.1477625
  • Baell, J. B., & Holloway, G. A. (2010). New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. Journal of Medicinal Chemistry, 53(7), 2719–2740. doi:10.1021/jm901137j
  • Banerjee, S., Adhikari, N., Amin, S. A., & Jha, T. (2020). Structural exploration of tetrahydroisoquinoline derivatives as HDAC8 inhibitors through multi-QSAR modeling study. Journal of Biomolecular Structure and Dynamics, 38(5), 1514–1551. doi:10.1080/07391102.2019.1617782
  • Banerjee, S., Amin, S., Abdul, Adhikari, N., & Jha, T. (2019). Essential elements regulating HDAC8 inhibition: A classification based structural analysis and enzyme-inhibitor interaction study of hydroxamate based HDAC8 inhibitors. Journal of Biomolecular Structure and Dynamics, 1–13. doi:10.1080/07391102.2019.1704881
  • Bergman, J. A., Woan, K., Perez-Villarroel, P., Villagra, A., Sotomayor, E. M., & Kozikowski, A. P. (2012). Selective histone deacetylase 6 inhibitors bearing substituted urea linkers inhibit melanoma cell growth. Journal of Medicinal Chemistry, 55(22), 9891–9899. doi:10.1021/jm301098e
  • Butler, K. V., Kalin, J., Brochier, C., Vistoli, G., Langley, B., & Kozikowski, A. P. (2010). Rational design and simple chemistry yield a superior, neuroprotective HDAC6 inhibitor, tubastatin A. Journal of the American Chemical Society, 132(31), 10842–10846. doi:10.1021/ja102758v
  • Case, D. A., Cheatham, T. E., Darden, T., Gohlke, H., Luo, R., Merz, K. M., Onufriev, A., Simmerling, C., Wang, B., & Woods, R. J. (2005). The Amber biomolecular simulation programs. Journal of Computational Chemistry, 26(16), 1668–1688. doi:10.1002/jcc.20290
  • Cheng, C., Yun, F., He, J., Ullah, S., & Yuan, Q. (2019). Design, synthesis and biological evaluation of novel thioquinazolinone-based 2-aminobenzamide derivatives as potent histone deacetylase (HDAC) inhibitors. European Journal of Medicinal Chemistry., 173, 185–202. doi:10.1016/j.ejmech.2019.04.017
  • De Vreese, R., Galle, L., Depetter, Y., Franceus, J., Desmet, T., Van Hecke, K., Benoy, V., Van Den Bosch, L., & D'hooghe, M. (2017). Synthesis of Potent and Selective HDAC6 Inhibitors Bearing a Cyclohexane- or Cycloheptane-Annulated 1,5-Benzothiazepine Scaffold. Chemistry - A European Journal, 23(1), 128–136. doi:10.1002/chem.201604167
  • Dewaker, V., Srivastava, P. N., Verma, S., & Prabhakar, Y. S. (2020). Molecular dynamics study of HDAC8-largazole analogues co-crystals for designing potential anticancer compounds. Journal of Biomolecular Structure and Dynamics, 38(4), 1117–1197. doi:10.1080/07391102.2019.1598497
  • Fournel, M., Bonfils, C., Hou, Y., Yan, P. T., Trachy-Bourget, M.-C., Kalita, A., Liu, J., Lu, A.-H., Zhou, N. Z., Robert, M.-F., Gillespie, J., Wang, J. J., Ste-Croix, H., Rahil, J., Lefebvre, S., Moradei, O., Delorme, D., MacLeod, A. R., Besterman, J. M., & Li, Z. (2008). MGCD0103, a novel isotype-selective histone deacetylase inhibitor, has broad spectrum antitumor activity in vitro and in vivo. Molecular Cancer Therapeutics, 7(4), 759–768. doi:10.1158/1535-7163.MCT-07-2026
  • Furumai, R., Matsuyama, A., Kobashi, N., Lee, K. H., Nishiyama, M., Nakajima, H., Tanaka, A., Komatsu, Y., Nishino, N., Yoshida, M., & Horinouchi, S. (2002). FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Research, 62(17), 4916–4921.
  • Grozinger, C. M., & Schreiber, S. L. (2002). Deacetylase enzymes: Biological functions and the use of small-molecule inhibitors. Chemistry & Biology, 9(1), 3–16. doi:10.1016/S1074-5521(02)00092-3
  • Hadley, M., Noonepalle, S., Banik, D., & Villagra, A. (2019). Functional analysis of HDACs in tumorigenesis. Methods in Molecular Biology, 1983, 279–307. doi:10.1007/978-1-4939-9434-2_17
  • Hai, Y., & Christianson, D. W. (2016). Histone deacetylase 6 structure and molecular basis of catalysis and inhibition. Nature Chemical Biology, 12(9), 741–747. doi:10.1038/nchembio.2134
  • Hermant, P., Bosc, D., Piveteau, C., Gealageas, R., Lam, B. V. y., Ronco, C., Roignant, M., Tolojanahary, H., Jean, L., Renard, P.-Y., Lemdani, M., Bourotte, M., Herledan, A., Bedart, C., Biela, A., Leroux, F., Deprez, B., & Deprez-Poulain, R. (2017). Controlling plasma stability of hydroxamic acids: A MedChem Toolbox. Journal of Medicinal Chemistry, 60(21), 9067–9089. doi:10.1021/acs.jmedchem.7b01444
  • Hrzenjak, A., Moinfar, F., Kremser, M. L., Strohmeier, B., Petru, E., Zatloukal, K., & Denk, H. (2010). Histone deacetylase inhibitor vorinostat suppresses the growth of uterine sarcomas in vitro and in vivo. Molecular Cancer, 9(1), 49. doi:10.1186/1476-4598-9-49
  • Humphrey, W., Dalke, A., & Schulten, K. (1996). VMD: Visual molecular dynamics. Journal of Molecular Graphics, 14(1), 33–38. doi:10.1016/0263-7855(96)00018-5
  • Ibrahim Uba, A., & Yelekci, K. (2019). Homology modeling of human histone deacetylase 10 and design of potential selective inhibitors. Journal of Biomolecular Structure and Dynamics, 37(14), 3627–3636. doi:10.1080/07391102.2018.1521747
  • Inks, E. S., Josey, B. J., Jesinkey, S. R., & Chou, C. J. (2012). A novel class of small molecule inhibitors of HDAC6. ACS Chemical Biology, 7(2), 331–339. doi:10.1021/cb200134p
  • Jain, A. N. (2008). Bias, reporting, and sharing: Computational evaluations of docking methods. Journal of Computer-Aided Molecular Design, 22(3–4), 201–212. doi:10.1007/s10822-007-9151-x
  • Kashyap, K., & Kakkar, R. (2020a). Exploring structural requirements of isoform selective histone deacetylase inhibitors: A comparative in silico study. Journal of Biomolecular Structure and Dynamics, 1–16. doi:10.1080/07391102.2019.1711191
  • Kashyap, K., & Kakkar, R. (2020b). An insight into selective and potent inhibition of histone deacetylase 8 through induced-fit docking, pharmacophore modeling and QSAR studies. Journal of Biomolecular Structure and Dynamics, 38(1), 48–65. doi:10.1080/07391102.2019.1567388
  • Kozikowski, A. P., Shen, S., Pardo, M., Tavares, M. T., Szarics, D., Benoy, V., Zimprich, C. A., Kutil, Z., Zhang, G., Bařinka, C., Robers, M. B., Van Den Bosch, L., Eubanks, J. H., & Jope, R. S. (2019). Brain penetrable histone deacetylase 6 inhibitor SW-100 ameliorates memory and learning impairments in a mouse model of fragile X syndrome. ACS Chemical Neuroscience, 10(3), 1679–1695. doi:10.1021/acschemneuro.8b00600
  • Krishna, S., Lakra, A. D., Shukla, N., Khan, S., Mishra, D. P., Ahmed, S., & Siddiqi, M. I. (2019). Identification of potential histone deacetylase1 (HDAC1) inhibitors using multistep virtual screening approach including SVM model, pharmacophore modeling, molecular docking and biological evaluation. Journal of Biomolecular Structure and Dynamics, 1–16. doi:10.1080/07391102.2019.1654925
  • Krukowski, K., Ma, J., Golonzhka, O., Laumet, G. O., Gutti, T., van Duzer, J. H., Mazitschek, R., Jarpe, M. B., Heijnen, C. J., & Kavelaars, A. (2017). HDAC6 inhibition effectively reverses chemotherapy-induced peripheral neuropathy. Pain, 158(6), 1126–1137. doi:10.1097/j.pain.0000000000000893
  • Lee, J.-H., Mahendran, A., Yao, Y., Ngo, L., Venta-Perez, G., Choy, M. L., Kim, N., Ham, W.-S., Breslow, R., & Marks, P. A. (2013). Development of a histone deacetylase 6 inhibitor and its biological effects. Proceedings of the National Academy of Sciences, 110(39), 15704–15709. doi:10.1073/pnas.1313893110
  • Li, Y., & Seto, E. (2016). HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harbor Perspectives in Medicine, 6(10), a026831. doi:10.1101/cshperspect
  • Lipinski, C. A. (2016). Rule of five in 2015 and beyond: Target and ligand structural limitations, ligand chemistry structure and drug discovery project decisions. Advanced Drug Delivery Reviews, 101, 34–41. doi:10.1016/j.addr.2016.04.029
  • Liu, J., Zhu, Y., He, Y., Zhu, H., Gao, Y., Li, Z., Zhu, J., Sun, X., Fang, F., Wen, H., & Li, W. (2020). Combined pharmacophore modeling, 3D-QSAR and docking studies to identify novel HDAC inhibitors using drug repurposing. Journal of Biomolecular Structure and Dynamics, 38(2), 533–547. doi:10.1080/07391102.2019.1590241
  • Liu, J. R., Yu, C. W., Hung, P. Y., Hsin, L. W., & Chern, J. W. (2019). High-selective HDAC6 inhibitor promotes HDAC6 degradation following autophagy modulation and enhanced antitumor immunity in glioblastoma. Biochemical Pharmacology, 163, 458–471. doi:10.1016/j.bcp.2019.03.023
  • Ning, Z.-Q., Li, Z.-B., Newman, M. J., Shan, S., Wang, X.-H., Pan, D.-S., Zhang, J., Dong, M., Du, X., & Lu, X.-P. (2012). Chidamide (CS055/HBI-8000): A new histone deacetylase inhibitor of the benzamide class with antitumor activity and the ability to enhance immune cell-mediated tumor cell cytotoxicity. Cancer Chemotherapy and Pharmacology, 69(4), 901–909. doi:10.1007/s00280-011-1766-x
  • Plumb, J. A., Finn, P. W., Williams, R. J., Bandara, M. J., Romero, M. R., Watkins, C. J., La Thangue, N. B., & Brown, R. (2003). Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Molecular Cancer Therapeutics, 2(8), 721–728.
  • Qin, H. T., Li, H. Q., & Liu, F. (2017). Selective histone deacetylase small molecule inhibitors: Recent progress and perspectives. Expert Opinion on Therapeutic Patents, 27(5), 621–636. doi:10.1080/13543776.2017.1276565
  • Saito, A., Yamashita, T., Mariko, Y., Nosaka, Y., Tsuchiya, K., Ando, T., Suzuki, T., Tsuruo, T., & Nakanishi, O. (1999). A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proceedings of the National Academy of Sciences, 96(8), 4592–4597. doi:10.1073/pnas.96.8.4592
  • Scuto, A., Kirschbaum, M., Kowolik, C., Kretzner, L., Juhasz, A., Atadja, P., Pullarkat, V., Bhatia, R., Forman, S., Yen, Y., & Jove, R. (2008). The novel histone deacetylase inhibitor, LBH589, induces expression of DNA damage response genes and apoptosis in Ph- acute lymphoblastic leukemia cells. Blood, 111(10), 5093–5100. doi:10.1182/blood-2007-10-117762
  • Senger, J., Melesina, J., Marek, M., Romier, C., Oehme, I., Witt, O., Sippl, W., & Jung, M. (2016). Synthesis and biological investigation of oxazole hydroxamates as highly selective histone deacetylase 6 (HDAC6) inhibitors. Journal of Medicinal Chemistry, 59(4), 1545–1555. doi:10.1021/acs.jmedchem.5b01493
  • Sharma, M., Jha, P., Verma, P., & Chopra, M. (2019). Combined comparative molecular field analysis, comparative molecular similarity indices analysis, molecular docking and molecular dynamics studies of histone deacetylase 6 inhibitors. Chemical Biology & Drug Design, 93(5), 910–925. doi:10.1111/cbdd.13488
  • Sixto-Lopez, Y., Bello, M., & Correa-Basurto, J. (2018). Structural and energetic basis for the inhibitory selectivity of both catalytic domains of dimeric HDAC6. Journal of Biomolecular Structure and Dynamics, 37 (18), 4701–4720. doi:10.1080/07391102.2018.1557560
  • Sixto-Lopez, Y., Bello, M., & Correa-Basurto, J. (2019). Insights into structural features of HDAC1 and its selectivity inhibition elucidated by Molecular dynamic simulation and Molecular Docking. Journal of Biomolecular Structure and Dynamics, 37(3), 584–610. doi:10.1080/07391102.2018.1441072
  • Subramanian, S., Bates, S. E., Wright, J. J., Espinoza-Delgado, I., & Piekarz, R. L. (2010). Clinical toxicities of histone deacetylase inhibitors. Pharmaceuticals (Basel), 3(9), 2751–2767. doi:10.3390/ph3092751
  • Sun, J.-y., Wang, J.-d., Wang, X., Liu, H.-c., Zhang, M.-m., Liu, Y.-C., Zhang, C.-h., Su, Y., Shen, Y.-y., Guo, Y.-w., Shen, A.-j., & Geng, M.-y. (2017). Marine-derived chromopeptide A, a novel class I HDAC inhibitor, suppresses human prostate cancer cell proliferation and migration. Acta Pharmacologica Sinica, 38(4), 551–560. doi:10.1038/aps.2016.139
  • Trott, O., & Olson, A. J. (2009). AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of Computational Chemistry, 31(2), 455–461. doi:10.1002/jcc.21334
  • Uba, A. I., Weako, J., Keskin, O., Gursoy, A., & Yelekci, K. (2019). Examining the stability of binding modes of the co-crystallized inhibitors of human HDAC8 by molecular dynamics simulation. Journal of Biomolecular Structure and Dynamics, 38(6), 1751-1760. doi:10.1080/07391102.2019.1615989
  • Uba, A. I., & Yelekci, K. (2018). Pharmacophore-based virtual screening for identification of potential selective inhibitors of human histone deacetylase 6. Computational Biology and Chemistry, 77, 318–330. doi:10.1016/j.compbiolchem.2018.10.016
  • Uba, A. I., & Yelekci, K. (2019). Crystallographic structure versus homology model: A case study of molecular dynamics simulation of human and zebrafish histone deacetylase 10. Journal of Biomolecular Structure and Dynamics, 1–12. doi:10.1080/07391102.2019.1691658
  • Vergani, B., Caprini, G., Fossati, G., Lattanzio, M., Marchini, M., Pavich, G., Pezzuto, M., Ripamonti, C., Sandrone, G., Steinkühler, C., & Stevenazzi, A. (2018). Selective HDAC6 inhibitors. WO2018189340A1.
  • Vergani, B., Sandrone, G., Marchini, M., Ripamonti, C., Cellupica, E., Galbiati, E., Caprini, G., Pavich, G., Porro, G., Rocchio, I., Lattanzio, M., Pezzuto, M., Skorupska, M., Cordella, P., Pagani, P., Pozzi, P., Pomarico, R., Modena, D., Leoni, F., … Stevenazzi, A. (2019). Novel benzohydroxamate-based potent and selective histone deacetylase 6 (HDAC6) inhibitors bearing a pentaheterocyclic scaffold: design, synthesis, and biological evaluation. Journal of Medicinal Chemistry, 62(23), 10711–10739. doi:10.1021/acs.jmedchem.9b01194
  • Vigushin, D. M., Mirsaidi, N., Brooke, G., Sun, C., Pace, P., Inman, L., Moody, C. J., & Coombes, R. C. (2004). Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Medical Oncology, 21 (1), 21–976. doi:10.1385/MO:21:1:21
  • Walji, A., Berger, R., Stump, C. A., Schlegel, K., Ann, S., Mulhearn, J. J., Greshock, T. J., Wang, D., Fraley, M. E., & Jones, K. G. (2017). Preparation of 3-aryl and heteroaryl substituted 5-trifluoromethyl oxadiazoles as histone deacetylase 6 (HDAC6) inhibitors. WO2017222951A1.
  • Wang, P., Wang, Z., & Liu, J. (2020). Role of HDACs in normal and malignant hematopoiesis. Molecular Cancer, 19(1), 5. doi:10.1186/s12943-019-1127-7
  • Wang, X. X., Wan, R. Z., & Liu, Z. P. (2018). Recent advances in the discovery of potent and selective HDAC6 inhibitors. European Journal of Medicinal Chemistry, 143, 1406–1418. doi:10.1016/j.ejmech.2017.10.040
  • Wang, Y., Yang, L., Hou, J., Zou, Q., Gao, Q., Yao, W., Yao, Q., & Zhang, J. (2019). Hierarchical virtual screening of the dual MMP-2/HDAC-6 inhibitors from natural products based on pharmacophore models and molecular docking. Journal of Biomolecular Structure and Dynamics, 37(3), 649–670. doi:10.1080/07391102.2018.1434833
  • Yuan, Y., Hu, Z., Bao, M., Sun, R., Long, X., Long, L., Li, J., Wu, C., & Bao, J. (2019). Screening of novel histone deacetylase 7 inhibitors through molecular docking followed by a combination of molecular dynamics simulations and ligand-based approach. Journal of Biomolecular Structure and Dynamics, 37(15), 4092–4103. doi:10.1080/07391102.2018.1541141
  • Zeb, A., Park, C., Rampogu, S., Son, M., Lee, G., & Lee, K. W. (2019). Structure-based drug designing recommends HDAC6 inhibitors to attenuate microtubule-associated tau-pathogenesis. ACS Chemical Neuroscience, 10(3), 1326–1335. doi:10.1021/acschemneuro.8b00405
  • Zeb, A., Park, C., Son, M., Rampogu, S., Alam, S. I., Park, S. J., & Lee, K. W. (2018). Investigation of non-hydroxamate scaffolds against HDAC6 inhibition: A pharmacophore modeling, molecular docking, and molecular dynamics simulation approach. Journal of Bioinformatics and Computational Biology, 16(03), 1840015. doi:10.1142/S0219720018400152
  • Zhang, X., Yuan, Z., Zhang, Y., Yong, S., Salas-Burgos, A., Koomen, J., Olashaw, N., Parsons, J. T., Yang, X.-J., Dent, S. R., Yao, T.-P., Lane, W. S., & Seto, E. (2007). HDAC6 modulates cell motility by altering the acetylation level of cortactin. Molecular Cell, 27(2), 197–213. doi:10.1016/j.molcel.2007.05.033

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.