IN-SILICO STRATEGIES IN ALZHEIMER’S DRUG DISCOVERY: MOLECULAR DOCKING, ADMET, AND PHARMACOLOGICAL PROFILING OF NOVEL HYDRAZIDE DERIVATIVES
Main Article Content
Keywords
Alzheimer’s disease, hydrazide derivatives, molecular docking, ADMET, pharmacokinetics, acetylcholinesterase, butyrylcholinesterase, in-silico drug design, neurodegenerative disorders, drug-likeness
Abstract
Background:
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder marked by memory loss, cognitive decline, and behavioral changes, primarily affecting the elderly population. It is histopathologically characterized by the accumulation of β-amyloid plaques and neurofibrillary tangles composed of hyperphosphorylated tau proteins. Despite extensive research, therapeutic options for AD remain limited, often only alleviating symptoms without halting disease progression. (1) The urgent need for disease-modifying agents has prompted researchers to explore novel compounds through computational approaches. Among emerging pharmacophores, hydrazide derivatives have garnered attention due to their wide-ranging biological activities, including neuroprotective and anti-oxidative properties. Leveraging in-silico techniques such as molecular docking and ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) profiling enables cost-effective and efficient screening of these compounds for potential anti-Alzheimer’s activity.
Objective: This study aims to evaluate the therapeutic potential of newly designed hydrazide derivatives as inhibitors of key enzymes involved in Alzheimer’s pathology, particularly acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). The research focuses on identifying promising lead candidates by assessing their binding affinities, drug-likeness, and pharmacokinetic properties using in-silico methodologies.
Methods: A library of novel hydrazide derivatives was designed based on structure-activity relationship (SAR) insights. Molecular docking simulations were conducted using AutoDock Vina to predict the binding affinities and interaction profiles of the compounds with AChE and BChE. The crystal structures of the target enzymes were obtained from the Protein Data Bank (PDB), and protein-ligand interactions were visualized using tools like Discovery Studio and PyMOL. ADMET profiling was performed using SwissADME and pkCSM to evaluate the drug-likeness, oral bioavailability, and safety parameters of the ligands. Additionally, Lipinski’s Rule of Five and Veber’s rules were applied to determine pharmacokinetic feasibility.
Results: Several hydrazide derivatives demonstrated strong binding affinity towards AChE and BChE, with docking scores indicating favorable interactions within the active site residues, including π-π stacking, hydrogen bonding, and hydrophobic contacts. The most promising candidates exhibited binding energies comparable to or better than standard inhibitors such as donepezil and rivastigmine. ADMET analysis revealed that the top hits possessed high gastrointestinal absorption, low blood-brain barrier permeability (selectively beneficial in reducing peripheral side effects), and minimal predicted hepatotoxicity or cardiotoxicity. Furthermore, the majority of the compounds adhered to Lipinski’s and Veber’s rules, suggesting good oral bioavailability and drug-likeness.
Conclusions: The in-silico evaluation of hydrazide derivatives presents a promising avenue for the development of novel anti-Alzheimer’s agents. The integration of molecular docking and ADMET profiling enabled the identification of potential leads with favorable interaction profiles and acceptable pharmacokinetic properties. While these findings provide a solid foundation, experimental validation through in-vitro and in-vivo studies is essential to confirm the therapeutic efficacy and safety of the shortlisted compounds. This computational strategy not only accelerates the early stages of drug discovery but also reduces the reliance on time-consuming and costly laboratory procedures.
References
2. Awasthi, S., & Mishra, A. (2020). Role of hydrazide derivatives in neurodegenerative diseases: A review. European Journal of Medicinal Chemistry, 199, 112384. https://doi.org/10.1016/j.ejmech.2020.112384
3. Berman, H. M., Westbrook, J., Feng, Z., et al. (2000). The Protein Data Bank. Nucleic Acids Research, 28(1), 235–242. https://doi.org/10.1093/nar/28.1.235
4. Chen, H., & Zhao, W. (2022). Advances in molecular docking for drug discovery targeting Alzheimer's disease. Frontiers in Pharmacology, 13, 789012. https://doi.org/10.3389/fphar.2022.789012
5. Chou, K. C., & Wei, D. Q. (2019). Computational drug design for Alzheimer's disease. Chemical Reviews, 119(2), 1314–1340. https://doi.org/10.1021/acs.chemrev.8b00395
6. Dassault Systèmes BIOVIA. (2021). Discovery Studio Modeling Environment, Release 2021. San Diego: Dassault Systèmes.
7. De Moraes, J., & Souza, M. (2020). Hydrazide derivatives: Versatile pharmacophores in medicinal chemistry. European Journal of Medicinal Chemistry, 198, 112357. https://doi.org/10.1016/j.ejmech.2020.112357
8. Daina, A., Michielin, O., & Zoete, V. (2017). SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness, and medicinal chemistry friendliness of small molecules. Scientific Reports, 7, 42717. https://doi.org/10.1038/srep42717
9. Dong, X., & Zhang, J. (2021). In-silico screening and ADMET prediction of novel AChE inhibitors. Journal of Molecular Liquids, 334, 116182. https://doi.org/10.1016/j.molliq.2021.116182
10. Du, X., & Zhao, H. (2019). Molecular dynamics simulations and free energy calculations in Alzheimer's drug discovery. Frontiers in Chemistry, 7, 712. https://doi.org/10.3389/fchem.2019.00712
11. Ekins, S., & Williams, A. J. (2018). Reproducibility and reuse of docking protocols: A call for standardization. Drug Discovery Today, 23(7), 1299–1302. https://doi.org/10.1016/j.drudis.2018.03.004
12. Ferraz, A. C., Gomes, R. A., & Ferreira, M. J. U. (2020). Design and synthesis of hydrazide derivatives as multifunctional anti-Alzheimer agents. Bioorganic Chemistry, 99, 103832. https://doi.org/10.1016/j.bioorg.2020.103832
13. Gaulton, A., Hersey, A., Nowotka, M., et al. (2017). The ChEMBL database in 2017. Nucleic Acids Research, 45(D1), D945–D954. https://doi.org/10.1093/nar/gkw1074
14. Guedes, I. A., de Magalhães, C. S., & Dardenne, L. E. (2014). Receptor–ligand molecular docking. Biophysical Reviews, 6(1), 75–87. https://doi.org/10.1007/s12551-013-0127-1
15. Gupta, A., & Pathak, C. (2019). Hydrazide and hydrazone derivatives: Potential molecules for Alzheimer's disease treatment. Journal of Enzyme Inhibition and Medicinal Chemistry, 34(1), 1209–1222. https://doi.org/10.1080/14756366.2019.1627894
16. Hall, M. L., & Rao, P. (2021). ADMET optimization in CNS drug discovery: Challenges and strategies. Expert Opinion on Drug Metabolism & Toxicology, 17(9), 1035–1047. https://doi.org/10.1080/17425255.2021.1896597
17. Han, L., & Wu, Y. (2023). Machine learning approaches for ADMET prediction in neurodegenerative diseases. Briefings in Bioinformatics, 24(2), bbac582. https://doi.org/10.1093/bib/bbac582
18. Huang, N., Shoichet, B. K., & Irwin, J. J. (2006). Benchmarking sets for molecular docking. Journal of Medicinal Chemistry, 49(23), 6789–6801. https://doi.org/10.1021/jm0608356
19. Jain, A. N. (2003). Surflex: Fully automatic flexible molecular docking using a molecular similarity-based search engine. Journal of Medicinal Chemistry, 46(4), 499–511. https://doi.org/10.1021/jm020406h
20. Jorgensen, W. L. (2004). The many roles of computation in drug discovery. Science, 303(5665), 1813–1818. https://doi.org/10.1126/science.1096361
21. Kabsch, W. (2010). XDS. Acta Crystallographica Section D: Biological Crystallography, 66(2), 125–132. https://doi.org/10.1107/S0907444909047337
22. Khandelwal, A., & Das, S. (2019). Design and development of multi-target hydrazide hybrids for Alzheimer's therapy. Bioorganic & Medicinal Chemistry Letters, 29(5), 613–620. https://doi.org/10.1016/j.bmcl.2018.12.048
23. Kim, S., Chen, J., Cheng, T., et al. (2019). PubChem 2019 update: Improved access to chemical data. Nucleic Acids Research, 47(D1), D1102–D1109. https://doi.org/10.1093/nar/gky1033
24. Kola, I., & Landis, J. (2004). Can the pharmaceutical industry reduce attrition rates? Nature Reviews Drug Discovery, 3(8), 711–715. https://doi.org/10.1038/nrd1470
25. Kumar, A., & Singh, A. (2021). Hydrazide derivatives as multifunctional agents in neurodegenerative disorders. European Journal of Medicinal Chemistry, 219, 113442. https://doi.org/10.1016/j.ejmech.2021.113442
26. Landrum, G. (2016). RDKit: Open-source cheminformatics. http://www.rdkit.org
27. Lee, M. S., & Olson, M. A. (2006). Calculation of binding free energies using molecular dynamics and free energy perturbation. Chemical Reviews, 106(5), 1801–1822. https://doi.org/10.1021/cr050304s
28. Li, J., & Fu, Z. (2018). Hydrazide-based inhibitors: Recent advances in design and applications. Medicinal Research Reviews, 38(5), 1612–1635. https://doi.org/10.1002/med.21456
29. Liu, X., Ouyang, S., Yu, B., et al. (2019). PharmMapper server: A web server for potential drug target identification using pharmacophore mapping approach. Nucleic Acids Research, 38(suppl_2), W609–W614. https://doi.org/10.1093/nar/gkq300
30. Macalino, S. J. Y., Gosu, V., Hong, S., & Choi, S. (2015). Role of computer-aided drug design in modern drug discovery. Archives of Pharmacal Research, 38(9), 1686–1701. https://doi.org/10.1007/s12272-015-0640-8
31. Majeed, S., & Raza, S. (2020). ADMET profiling in CNS drug discovery: The importance of BBB permeability prediction. Frontiers in Pharmacology, 11, 1073. https://doi.org/10.3389/fphar.2020.01073
32. Manoharan, P. T., & Sriram, D. (2017). Hydrazide-based scaffolds: Pharmacological and therapeutic potential. European Journal of Medicinal Chemistry, 126, 251–268. https://doi.org/10.1016/j.ejmech.2016.11.054
33. Martins, D., & Outeiro, T. F. (2020). Tau aggregation and Alzheimer's disease: Molecular docking studies towards novel inhibitors. Molecules, 25(16), 3669. https://doi.org/10.3390/molecules25163669
34. McGann, M. (2011). FRED pose prediction and virtual screening accuracy. Journal of Chemical Information and Modeling, 51(3), 578–596. https://doi.org/10.1021/ci100436p
35. McQuaid, T. J., & Bush, A. I. (2021). Metal chelation therapy and Alzheimer's disease. Neurotherapeutics, 18(3), 1236–1250. https://doi.org/10.1007/s13311-021-01087-8
36. Morris, G. M., Huey, R., Lindstrom, W., et al. (2009). AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of Computational Chemistry, 30(16), 2785–2791. https://doi.org/10.1002/jcc.21256
37. Mu, Y., & Zhang, H. (2019). Recent advances in BACE1 inhibitors: Design, structure–activity relationships and biological evaluation. European Journal of Medicinal Chemistry, 168, 1–23. https://doi.org/10.1016/j.ejmech.2019.01.004
38. Neves, B. J., & Vasconcelos, I. M. (2018). Molecular docking and ADMET studies for Alzheimer's drug discovery. Bioorganic Chemistry, 78, 52–61. https://doi.org/10.1016/j.bioorg.2018.05.008
39. Ochoa, V., & Pascual, M. (2020). Role of pharmacokinetics in CNS drug discovery. Drug Metabolism Reviews, 52(4), 383–395. https://doi.org/10.1080/03602532.2020.1808297
40. Olson, R., & Smith, J. (2017). The importance of standardization in docking studies. Journal of Computer-Aided Molecular Design, 31(6), 549–561. https://doi.org/10.1007/s10822-017-0036-2
41. Panda, S., & Verma, A. (2021). ADMET optimization in drug discovery: Applications and challenges. Current Drug Metabolism, 22(12), 958–971. https://doi.org/10.2174/1389200222666210415121021
42. Parrinello, M., & Rahman, A. (1981). Polymorphic transitions in single crystals: A new molecular dynamics method. Journal of Applied Physics, 52(12), 7182–7190. https://doi.org/10.1063/1.328693
43. Patel, H., & Patel, A. (2019). Hydrazide derivatives as potential multifunctional anti-Alzheimer's agents. Journal of Enzyme Inhibition and Medicinal Chemistry, 34(1), 134–146. https://doi.org/10.1080/14756366.2018.1545763
44. Pires, D. E., Blundell, T. L., & Ascher, D. B. (2015). pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. Journal of Medicinal Chemistry, 58(9), 4066–4072. https://doi.org/10.1021/acs.jmedchem.5b00104
45. Ramsay, R. R., & Tipton, K. F. (2017). The structural basis of monoamine oxidase inhibition by hydrazines and hydrazides. Journal of Neural Transmission, 124(6), 671–682. https://doi.org/10.1007/s00702-017-1704-3
46. Rashid, N., & Lee, S. J. (2020). Systems pharmacology approaches in Alzheimer's disease drug discovery. Frontiers in Pharmacology, 11, 819. https://doi.org/10.3389/fphar.2020.00819
47. Roy, A., & Kundu, S. (2018). Molecular docking and pharmacophore modeling of hydrazide derivatives as acetylcholinesterase inhibitors. Journal of Molecular Structure, 1169, 145–154. https://doi.org/10.1016/j.molstruc.2018.05.030
48. Sadowski, J., Gasteiger, J., & Klebe, G. (1994). Comparison of automatic three-dimensional model builders using 639 X-ray structures. Journal of Chemical Information and Computer Sciences, 34(4), 1000–1008. https://doi.org/10.1021/ci00021a033
49. Sakkiah, S., Thangapandian, S., John, S., et al. (2012). Molecular docking and molecular dynamics studies of acetylcholinesterase inhibitors. Journal of Molecular Modeling, 18(5), 1993–2005. https://doi.org/10.1007/s00894-011-1274-1
50. Schrödinger Release 2023-1: Glide, Schrödinger, LLC, New York, NY, 2023.
51. Shaikh, S. A., & Kumar, P. (2022). Design, synthesis, and biological evaluation of hydrazide derivatives as multifunctional neuroprotective agents. *European Journal of Medicinal Chemistry, 235
52. Singh, P., & Verma, A. (2021). Molecular docking and ADMET studies of novel hydrazide derivatives as potential anti-Alzheimer agents. Journal of Biomolecular Structure and Dynamics, 39(7), 2468–2478. https://doi.org/10.1080/07391102.2020.1756954
53. Sousa, S. F., Fernandes, P. A., & Ramos, M. J. (2006). Protein–ligand docking: Current status and future challenges. Proteins: Structure, Function, and Bioinformatics, 65(1), 15–26. https://doi.org/10.1002/prot.21082
54. Sun, L., & Li, H. (2020). Computational approaches for ADMET prediction in drug discovery. Current Topics in Medicinal Chemistry, 20(21), 1887–1899. https://doi.org/10.2174/1568026620666200312102220
55. Trott, O., & Olson, A. J. (2010). 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. https://doi.org/10.1002/jcc.21334
56. Udhayakumar, V., & Subramanian, V. (2018). Design and synthesis of hydrazide derivatives targeting acetylcholinesterase and butyrylcholinesterase for Alzheimer’s disease. Bioorganic Chemistry, 77, 561–568. https://doi.org/10.1016/j.bioorg.2018.05.015
57. Van der Spoel, D., Lindahl, E., Hess, B., et al. (2005). GROMACS: Fast, flexible, and free. Journal of Computational Chemistry, 26(16), 1701–1718. https://doi.org/10.1002/jcc.20291
58. Wang, J., Wang, W., Kollman, P. A., & Case, D. A. (2006). Automatic atom type and bond type perception in molecular mechanical calculations. Journal of Molecular Graphics and Modelling, 25(2), 247–260. https://doi.org/10.1016/j.jmgm.2005.12.005
59. Yang, S. Y. (2010). Pharmacophore modeling and applications in drug discovery: Challenges and recent advances. Drug Discovery Today, 15(11–12), 444–450. https://doi.org/10.1016/j.drudis.2010.03.013
60. Zhang, Y., & Skolnick, J. (2005). TM-align: A protein structure alignment algorithm based on the TM-score. Nucleic Acids Research, 33(7), 2302–2309. https://doi.org/10.1093/nar/gki524