Novel anthraquinone amino derivatives as anticancer agents targeting the human serine/threonine kinase PAK4

Author:

Hasan Hamad M.1,Koua Faisal H. M.2,Beit Hajer A.1,Ebrahim Reem M. A.3,Ismail Amar Mohamed4

Affiliation:

1. Omar Al-Mokhtar University

2. National University Biomedical Research Institute (NUBRI), National University- Sudan

3. African City of Technology

4. Al Neelain University

Abstract

Abstract

Recently, anthraquinone scaffolds have attracted increasing amounts of attention as sources for effective drug synthesis for the treatment of various diseases. Here, we demonstrate the potential of nine anthraquinone amino-derivatives (AADs) as anticancer agents. We synthesized nine Schiff bases (1-9) using an anthraquinone scaffold and amino acids. We applied in silico analysis by performing molecular docking against the anticancer drug target human serine/threonine p21-activated kinase 4 (PAK4) and compared them with the recently developed PAK4 inhibitor PF-3758309. Compounds 5and 8 showed docking properties comparable to that of PF-3758309 at two binding sites located in the C-terminal domain of PAK4. Compound 5, a tryptophan-based anthraquinone derivative, had docking scores of -9.3 and -8.7 kcal/mol at two different identified cavities, cav-1 and cav-4, respectively, exceeding that of the control PF-3758309 for both cavities. Additional optimization of the indole moiety of 5gave rise to 5b, resulting in high drug likeness (0.74) and comparable binding properties to cav-1 (-8.6 kcal/mol) and the highest achievable binding affinity to cav-4, with an average of -9.7 kcal/mol, indicating a potential allosteric inhibitory effect via cav-4. Our molecular dynamics simulation analysis of the top hits and the control compound provides important insights into the mechanism of action of these drug candidates. Additionally, all the compounds exhibited promising pharmacokinetic and toxicity properties, as revealed by the ADMET profiles, urging the need for comprehensive in vitro and in vivo antitumor characterization. Our results also indicate that cav-4 may serve as a target for designing specific allosteric inhibitors against PAK4-overexpressing cancer cells, suggesting its potential application in adjuvant cancer treatment.

Publisher

Research Square Platform LLC

Reference51 articles.

1. Targeting protein serine/threonine phosphatases for drug development;McConnell JL;Mol. Pharmacol.,2009

2. Structure, biochemistry, and biology of PAK kinases;Kumar R;Gene,2017

3. Kinase-targeted cancer therapies: progress, challenges and future directions;Bhullar KS;Mol. Cancer,2018

4. Bononi, A. et al. Protein kinases and phosphatases in the control of cell fate. Enzyme Res. 2011, 329098 (2011).

5. Effects of PAK4/LIMK1/Cofilin-1 signaling pathway on proliferation, invasion, and migration of human osteosarcoma cells;Li Z;J. Clin. Lab. Anal.,2020

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3