DOI QR코드

DOI QR Code

Pharmacological Analysis of Vorinostat Analogues as Potential Anti-tumor Agents Targeting Human Histone Deacetylases: an Epigenetic Treatment Stratagem for Cancers

  • Published : 2016.04.11

Abstract

Alteration of the acetylation status of chromatin and other non-histone proteins by HDAC inhibitors has evolved as an excellent epigenetic strategy in treatment of cancers. The present study was sought to identify compounds with positive pharmacological profiles targeting HDAC1. Analogues of Vorinostat synthesized by Cai et al, 2015 formed the test compounds for the present pharmacological evaluation. Hydroxamte analogue 6H showed superior pharmacological profile in comparison to all the compounds in the analogue dataset owing to its better electrostatic interactions and hydrogen bonding patterns. In order to identify compounds with even better high affinity and pharmacological profile than 6H and Vorinostat, virtual screening was performed. A total of 83 compounds similar to Vorinostat and 154 compounds akin to analogue 6H were retrieved. SCHEMBL15675695 (PubCid: 15739209) and AKOS019005527 (PubCid: 80442147) similar to Vorinostat and 6H, were the best docked compounds among the virtually screened compounds. However, in spite of having good affinity, none of the virtually screened compounds had better affinity than that of 6H. In addition SCHEMBL15675695 was predicted to be a carcinogen while AKOS019005527 is Ames toxic. From, our extensive analysis involving binding affinity analysis, ADMET properties predictions and pharmacophoric mappings, we report Vorinostat hydroxamate analogue 6H to be a potential candidate for HDAC inhibition in treatment of cancers through an epigenetic strategy.

Keywords

References

  1. Agrawal A, Murphy RF, Agrawal DK (2007). DNA methylation in breast and colorectal cancers. Mod Pathol, 20, 711-21 https://doi.org/10.1038/modpathol.3800822
  2. Bandaru S, Ponnala D, Lakkaraju C, et al (2014). Identification of high affinity non-peptidic small molecule inhibitors of mdm2-p53 interactions through structure-based virtual screening strategies. Asian Pac J Cancer Prev, 16, 3759-65.
  3. Bandaru S, Tiwari G, Akka J, et al (2015). Identification of high affinity bioactive salbutamol conformer directed against mutated (thr164ile) beta 2 adrenergic receptor. Curr Top Med Chem, 15, 50-6. https://doi.org/10.2174/1568026615666150112113040
  4. Blair LP, Yan Q (2012). Epigenetic mechanisms in commonly occurring cancers. DNA Cell Biol, 31, 49-61.
  5. Bolden JE, Peart MJ, Johnstone RW (2006). Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Disc, 5, 769-84 https://doi.org/10.1038/nrd2133
  6. Cai J, Wei H, Hong KH, et al (2015). Discovery, bioactivity and docking simulation of Vorinostat analogues containing 1, 2, 4-oxadiazole moiety as potent histone deacetylase inhibitors and antitumor agents. Bioorg Med Chem, 23, 3457-71. https://doi.org/10.1016/j.bmc.2015.04.028
  7. Cheng F, Li W, Zhou Y, et al (2012). Admet SAR: a comprehensive source and free tool for assessment of chemical ADMET properties. J Chem Inf Model, 52, 3099-105. https://doi.org/10.1021/ci300367a
  8. Clayton AL, Hazzalin CA, Mahadevan LC (2006). Enhanced histone acetylation and transcription: a dynamic perspective. Mol Cell, 23, 289-96. https://doi.org/10.1016/j.molcel.2006.06.017
  9. De Ruijter AJ, van Gennip AH, Caron HN, et al (2003). Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J, 370, 737-49 https://doi.org/10.1042/bj20021321
  10. Dokmanovic M, Clarke C, Marks PA (2007). Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res, 5, 981-9 https://doi.org/10.1158/1541-7786.MCR-07-0324
  11. Dokmanovic M, Marks PA (2005). Prospects: histone deacetylase inhibitors. J Cell Biochem, 96, 293-304 https://doi.org/10.1002/jcb.20532
  12. Fouladi M (2006). Histone deacetylase inhibitors in cancer therapy. Cancer Invest, 24, 521-7 https://doi.org/10.1080/07357900600814979
  13. Fraga MF, Ballestar E, Villar-Garea A, et al (2005). Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet, 37, 391-400 https://doi.org/10.1038/ng1531
  14. Glozak MA, Seto E (2007). Histone deacetylases and cancer. Oncogene, 26, 5420-32 https://doi.org/10.1038/sj.onc.1210610
  15. Grant S, Easley C, Kirkpatrick P (2007). Vorinostat. Nat Rev Drug Discov, 6, 21-2. https://doi.org/10.1038/nrd2227
  16. Johnstone RW (2002). Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov, 1, 287-99. https://doi.org/10.1038/nrd772
  17. Kelly TK, De Carvalho DD, Jones PA (2010). Epigenetic modifications as therapeutic targets. Nat Biotechnol, 28, 1069-78. https://doi.org/10.1038/nbt.1678
  18. Kelotra S, Jain M, Kelotra A, et al (2014). An in silico Appraisal to Identify High Affinity Anti-Apoptotic Synthetic Tetrapeptide Inhibitors Targeting the Mammalian Caspase 3 Enzyme. Asian Pac J Cancer Prev, 15, 10137-42.
  19. Lafon-Hughes L, Di Tomaso MV, Mendez-Acuna L, et al (2008). Chromatin-remodelling mechanisms in cancer. Mutat Res, 658, 191-214 https://doi.org/10.1016/j.mrrev.2008.01.008
  20. Lauffer, Benjamin EL, Robert Mintzer, et al (2013). Histone deacetylase (HDAC) inhibitor kinetic rate constants correlate with cellular histone acetylation but not transcription and cell viability. J Biol Chem, 288, 26926-43. https://doi.org/10.1074/jbc.M113.490706
  21. Magnaghi-Jaulin L, Eot-Houllier G, Fulcrand G, et al (2007). Histone deacetylase inhibitors induce premature sister chromatid separation and override the mitotic spindle assembly checkpoint. Cancer Res, 67, 6360-7 https://doi.org/10.1158/0008-5472.CAN-06-3012
  22. Mariadason JM (2008). HDACs and HDAC inhibitors in colon cancer. Epigenetics, 3, 28-37. https://doi.org/10.4161/epi.3.1.5736
  23. Nelder JA, Mead R (1965). A simplex method for function minimization. Comput J, 7, 308-13. https://doi.org/10.1093/comjnl/7.4.308
  24. Piekarz RL, Sackett DL, Bates SE (2007). Histone deacetylase inhibitors and demethylating agents: clinical development of histone deacetylase inhibitors for cancer therapy. Cancer J, 13, 30-9 https://doi.org/10.1097/PPO.0b013e31803c73cc
  25. Shen L, Issa JP (2002). Epigenetics in colorectal cancer. Curr Opin Gastroenterol, 18, 68-73 https://doi.org/10.1097/00001574-200201000-00012
  26. Stevens FE, Beamish H, Warrener R, et al (2008). Histone deacetylase inhibitors induce mitotic slippage. Oncogene, 27, 1345-54 https://doi.org/10.1038/sj.onc.1210779
  27. Thomsen R, Christensen MH (2006). MolDock: a new technique for high-accuracy molecular docking. J Med Chem, 49, 3315-21. https://doi.org/10.1021/jm051197e
  28. Xu WS, Perez G, Ngo L, et al (2005). Induction of polyploidy by histone deacetylase inhibitor: a pathway for antitumor effects. Cancer Res, 65, 7832-9 https://doi.org/10.1158/0008-5472.CAN-04-4608
  29. Yang JM, Chen CC (2004). Gemdock: A generic evolutionary method for molecular docking. Proteins, 55, 288-304. https://doi.org/10.1002/prot.20035