1
|
Giorgioni G, Bonifazi A, Botticelli L, Cifani C, Matteucci F, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Giannella M, Piergentili A, Piergentili A, Quaglia W, Del Bello F. Advances in drug design and therapeutic potential of selective or multitarget 5-HT1A receptor ligands. Med Res Rev 2024; 44:2640-2706. [PMID: 38808959 DOI: 10.1002/med.22049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/14/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024]
Abstract
5-HT1A receptor (5-HT1A-R) is a serotoninergic G-protein coupled receptor subtype which contributes to several physiological processes in both central nervous system and periphery. Despite being the first 5-HT-R identified, cloned and studied, it still represents a very attractive target in drug discovery and continues to be the focus of a myriad of drug discovery campaigns due to its involvement in numerous neuropsychiatric disorders. The structure-activity relationship studies (SAR) performed over the last years have been devoted to three main goals: (i) design and synthesis of 5-HT1A-R selective/preferential ligands; (ii) identification of 5-HT1A-R biased agonists, differentiating pre- versus post-synaptic agonism and signaling cellular mechanisms; (iii) development of multitarget compounds endowed with well-defined poly-pharmacological profiles targeting 5-HT1A-R along with other serotonin receptors, serotonin transporter (SERT), D2-like receptors and/or enzymes, such as acetylcholinesterase and phosphodiesterase, as a promising strategy for the management of complex psychiatric and neurodegenerative disorders. In this review, medicinal chemistry aspects of ligands acting as selective/preferential or multitarget 5-HT1A-R agonists and antagonists belonging to different chemotypes and developed in the last 7 years (2017-2023) have been discussed. The development of chemical and pharmacological 5-HT1A-R tools for molecular imaging have also been described. Finally, the pharmacological interest of 5-HT1A-R and the therapeutic potential of ligands targeting this receptor have been considered.
Collapse
Affiliation(s)
- Gianfabio Giorgioni
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
| | - Luca Botticelli
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Federica Matteucci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | | | | | - Mario Giannella
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | | | - Alessia Piergentili
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Wilma Quaglia
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Fabio Del Bello
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| |
Collapse
|
2
|
Mayo P, Pascual J, Crisman E, Domínguez C, López MG, León R. Innovative pathological network-based multitarget approaches for Alzheimer's disease treatment. Med Res Rev 2024; 44:2367-2419. [PMID: 38678582 DOI: 10.1002/med.22045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/02/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is a major health threat globally. Its prevalence is forecasted to exponentially increase during the next 30 years due to the global aging population. Currently, approved drugs are merely symptomatic, being ineffective in delaying or blocking the relentless disease advance. Intensive AD research describes this disease as a highly complex multifactorial disease. Disclosure of novel pathological pathways and their interconnections has had a major impact on medicinal chemistry drug development for AD over the last two decades. The complex network of pathological events involved in the onset of the disease has prompted the development of multitarget drugs. These chemical entities combine pharmacological activities toward two or more drug targets of interest. These multitarget-directed ligands are proposed to modify different nodes in the pathological network aiming to delay or even stop disease progression. Here, we review the multitarget drug development strategy for AD during the last decade.
Collapse
Affiliation(s)
- Paloma Mayo
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Jorge Pascual
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Cristina Domínguez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| |
Collapse
|
3
|
Ayoup MS, Barakat MR, Abdel-Hamid H, Emam E, Al-Faiyz YS, Masoud AA, Ghareeb DA, Sonousi A, Kassab AE. Design, synthesis, and biological evaluation of 1,2,4-oxadiazole-based derivatives as multitarget anti-Alzheimer agents. RSC Med Chem 2024; 15:2080-2097. [PMID: 38911158 PMCID: PMC11187554 DOI: 10.1039/d4md00113c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/04/2024] [Indexed: 06/25/2024] Open
Abstract
A series of novel 1,2,4-oxadiazole-based derivatives were synthesized and evaluated for their potential anti-Alzheimer disease activity. The results revealed that compounds 2b, 2c, 2d, 3a, 4a, 6, 9a, 9b, and 13b showed excellent inhibitory activity against acetylcholinesterase (AChE) with IC50 values in the range of 0.0158 to 0.121 μM. They were 1.01 to 7.78 times more potent than donepezil (IC50 = 0.123 μM). The newly synthesized compounds exhibited lower activity towards butyrylcholinesterase (BuChE) when compared to rivastigmine. Compounds 4b and 13b showed the most prominent inhibitory potential against BuChE with IC50 values of 11.50 and 15 μM, respectively. Moreover, 4b, and 9b were found to be more potent antioxidant agents (IC50 values of 59.25, and 56.69 μM, respectively) in comparison with ascorbic acid (IC50 = 74.55 μM). Compounds 2b and 2c exhibited monoamine oxidase-B (MAO-B) inhibitory activity with IC50 values of 74.68 and 225.48 μM, respectively. They were 3.55 and 1.17 times more potent than biperiden (IC50 = 265.85 μM). The prominent interactions of the compounds with the AChE active site can be used to computationally explain the high AChE inhibitory activity. The results unveiled 1,2,4-oxadiazole derivatives 2c and 3a as multitarget anti-AD agents. The predicted ADME properties for compounds 2b and 4a were satisfactory, and 4a had the highest likelihood of crossing the blood-brain barrier (BBB), making it the optimum compound for future optimization.
Collapse
Affiliation(s)
- Mohammed Salah Ayoup
- Department of Chemistry, College of Science, King Faisal University P.O. Box 400 Al-Ahsa 31982 Saudi Arabia
- Chemistry Department, Faculty of Science, Alexandria University P.O. Box 426 Alexandria 21321 Egypt
| | - Mohamed Reda Barakat
- Chemistry Department, Faculty of Science, Alexandria University P.O. Box 426 Alexandria 21321 Egypt
| | - Hamida Abdel-Hamid
- Chemistry Department, Faculty of Science, Alexandria University P.O. Box 426 Alexandria 21321 Egypt
| | - Ehab Emam
- General Q.C Manager, Alexandria company for pharmaceuticals Alexandria 21521 Egypt
| | - Yasair S Al-Faiyz
- Department of Chemistry, College of Science, King Faisal University P.O. Box 400 Al-Ahsa 31982 Saudi Arabia
| | - Aliaa A Masoud
- Bio-screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University 21511 Alexandria Egypt
| | - Doaa A Ghareeb
- Bio-screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University 21511 Alexandria Egypt
- Center of Excellence for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industry Development Center, City of Scientific Research & Technological Applications (SRTA-city) New Borg El Arab Alexandria Egypt
| | - Amr Sonousi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University P.O. Box 11562, Kasr El-Aini Street Cairo Egypt
- University of Hertfordshire hosted by Global Academic Foundation, New Administrative Capital Cairo Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University P.O. Box 11562, Kasr El-Aini Street Cairo Egypt
| |
Collapse
|
4
|
Qiu J, Feng X, Chen H, Liu W, Liu W, Wu L, Gao X, Liu Y, Huang Y, Gong H, Qi Y, Xu Z, Zhao Q. Discovery of novel harmine derivatives as GSK-3β/DYRK1A dual inhibitors for Alzheimer's disease treatment. Arch Pharm (Weinheim) 2024; 357:e2300404. [PMID: 38010470 DOI: 10.1002/ardp.202300404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/22/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023]
Abstract
Multitarget-directed ligands (MTDLs) have recently attracted significant interest due to their superior effectiveness in multifactorial Alzheimer's disease (AD). Combined inhibition of two important AD targets, glycogen synthase kinase-3β (GSK-3β) and dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), may be a breakthrough in the treatment of AD. Based on our previous work, we have designed and synthesized a series of novel harmine derivatives, investigated their inhibition of GSK-3β and DYRK1A, and evaluated a variety of biological activities. The results of the experiments showed that most of these compounds exhibited good activity against GSK-3β and DYRK1A in vitro. ZLQH-5 was selected as the best compound due to the most potent inhibitory effect against GSK-3β and DYRK1A. Molecular docking studies demonstrated that ZLQH-5 could form stable interactions with the ATP binding pocket of GSK-3β and DYRK1A. In addition, ZLQH-5 showed low cytotoxicity against SH-SY5Y and HL-7702, good blood-brain barrier permeability, and favorable pharmacokinetic properties. More importantly, ZLQH-5 also attenuated the tau hyperphosphorylation in the okadaic acid SH-SY5Y cell model. These results indicated that ZLQH-5 could be a promising dual-target drug candidate for the treatment of AD.
Collapse
Affiliation(s)
- Jingsong Qiu
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiangling Feng
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Huanhua Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Wenwu Liu
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
| | - Wenjie Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Limeng Wu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xudong Gao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Yanfang Liu
- Department of Clinical Trial Center, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaoguang Huang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Hao Gong
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yiming Qi
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Zihua Xu
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qingchun Zhao
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
5
|
Halder AK, Mitra S, Cordeiro MNDS. Designing multi-target drugs for the treatment of major depressive disorder. Expert Opin Drug Discov 2023; 18:643-658. [PMID: 37183604 DOI: 10.1080/17460441.2023.2214361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Major depressive disorders (MDD) pose major health burdens globally. Currently available medications have their limitations due to serious adverse effects, long latency periods as well as resistance. Considering the highly complicated pathological nature of this disorder, it has been suggested that multitarget drugs or multi-target-directed ligands (MTDLs) may provide long-term therapeutic solutions for the treatment of MDD. AREAS COVERED In the current review, recent lead design and lead modification strategies have been covered. Important investigations reported in the last ten years (2013-2022) for the pre-clinical development of MTDLs (through synthetic medicinal chemistry and biological evaluation) for the treatment of MDD were discussed as case studies to focus on the recent design strategies. The discussions are categorized based on the pharmacological targets. On the basis of these important case studies, the challenges involved in different design strategies were discussed in detail. EXPERT OPINION Even though large variations were observed in the selection of pharmacological targets, some potential biological targets (NMDA, melatonin receptors) are required to be explored extensively for the design of MTDLs. Similarly, apart from structure activity relationship (SAR), in silico techniques such as multitasking cheminformatic modelling, molecular dynamics simulation and virtual screening should be exploited to a greater extent.
Collapse
Affiliation(s)
- Amit Kumar Halder
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur 713206, India
| | - Soumya Mitra
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur 713206, India
| | - Maria Natalia D S Cordeiro
- LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| |
Collapse
|
6
|
Cong S, Shi Y, Yu G, Zhong F, Li J, Liu J, Ye C, Tan Z, Deng Y. Discovery of novel 5-(2-hydroxyphenyl)-2-phthalide-3(3H)-pyrazolones as balanced multifunctional agents against Alzheimer's disease. Eur J Med Chem 2023; 250:115216. [PMID: 36857812 DOI: 10.1016/j.ejmech.2023.115216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 02/21/2023]
Abstract
Based on previous work, a series of novel 5-(2-hydroxyphenyl)-2-phthalide-3(3H)-pyrazolones derivatives were identified as potential multifunctional therapeutic agents for Alzheimer's disease. Biological evaluation exhibited that these derivatives had great performance against MAO-B, Aβ1-42 aggregation, oxidative stress and metal ion dyshomeostasis. Among them, 10x was selected as the optimal agent for its excellent MAO-B inhibitory activity (IC50 = 0.41 μM, SI > 24.4), good antioxidant activity (1.16 Trolox equivalent) and anti-Aβ aggregation activity (56.03% and 57.51% for inhibition of self- and Cu2+-induced Aβ1-42 aggregation; 81.91% and 82.40% for disaggregation of self- and Cu2+-induced Aβ1-42 fibrils at 25.0 μM). Besides, 10x also exhibited obvious metal-ion chelating ability, anti-neuroinflammation (NO, TNF-α), neuroprotective activity and BBB permeability. More importantly, in vivo behavioral assessment demonstrated 10x could remarkably improve the memory and cognitive impairment in Aβ1-42 induced AD mice model. Overall, these test results indicated 10x could serve as a balanced multifunctional anti-AD agent and deserved further research.
Collapse
Affiliation(s)
- Shiqin Cong
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yichun Shi
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Guangjun Yu
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Feng Zhong
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jingjing Li
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jing Liu
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Chanyuan Ye
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhenghuai Tan
- Institute of Traditional Chinese Medicine Pharmacology and Toxicology, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Yong Deng
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Bubley A, Erofeev A, Gorelkin P, Beloglazkina E, Majouga A, Krasnovskaya O. Tacrine-Based Hybrids: Past, Present, and Future. Int J Mol Sci 2023; 24:ijms24021717. [PMID: 36675233 PMCID: PMC9863713 DOI: 10.3390/ijms24021717] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder which is characterized by β-amyloid (Aβ) aggregation, τ-hyperphosphorylation, and loss of cholinergic neurons. The other important hallmarks of AD are oxidative stress, metal dyshomeostasis, inflammation, and cell cycle dysregulation. Multiple therapeutic targets may be proposed for the development of anti-AD drugs, and the "one drug-multiple targets" strategy is of current interest. Tacrine (THA) was the first clinically approved cholinesterase (ChE) inhibitor, which was withdrawn due to high hepatotoxicity. However, its high potency in ChE inhibition, low molecular weight, and simple structure make THA a promising scaffold for developing multi-target agents. In this review, we summarized THA-based hybrids published from 2006 to 2022, thus providing an overview of strategies that have been used in drug design and approaches that have resulted in significant cognitive improvements and reduced hepatotoxicity.
Collapse
Affiliation(s)
- Anna Bubley
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
| | - Alexaner Erofeev
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Peter Gorelkin
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Elena Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
| | - Alexander Majouga
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
| | - Olga Krasnovskaya
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1-3, Moscow 119991, Russia
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, Moscow 119049, Russia
- Correspondence:
| |
Collapse
|
8
|
Wang J, Qian B, Wang T, Ma Y, Lin H, Zhang Y, Lv H, Zhang X, Hu Y, Xu S, Liu F, Li H, Jiang Z. Nontoxic Tb 3+-induced hyaluronic nano-poached egg aggregates for colorimetric and luminescent detection of Fe 3+ ions. RSC Adv 2022; 12:22285-22294. [PMID: 36043088 PMCID: PMC9366763 DOI: 10.1039/d2ra03871d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
This study demonstrates that a luminescent Tb3+ complex with green emission can be complexed with hyaluronic (hya) to form nanoparticles. The structure of complexation is composed of a Tb(acac)2phen core with a hya surface, similar to those of the nano-poached eggs. What makes the structure unique is that Tb(acac)2phen and hya are connected by chemical bonds. To confirm their utility, we illustrate that the luminescence is rapidly and selectively quenched in the presence of Fe3+. Initial cytotoxicity experiments with human liver carcinoma cells show that the luminescent lanthanide complexes are cytotoxic, however, complexing lanthanides to hya renders them cytocompatible. The new complex integrates the advantages of superior lanthanide luminescence, the unique shape of nano-poached eggs, compatibility with aqueous systems, and cytocompatibility. Tb3+-induced hyaluronic nano-poached eggs (THNE) can, therefore, be used for Fe3+ detection in aqueous systems. The original Tb3+-induced hyaluronic nano-poached eggs (THNE) integrates the advantages of superior lanthanide luminescence, the unique shape of nano-poached eggs, and non-toxicity, for the sensing of Fe3+ in aqueous surroundings.![]()
Collapse
Affiliation(s)
- Jing Wang
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Bei Qian
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University Qingdao 266109 China
| | - Tao Wang
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Yanyan Ma
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Haitao Lin
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Yimeng Zhang
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Hongmin Lv
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Xiaonan Zhang
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Yimeng Hu
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Shanshan Xu
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| | - Fengchen Liu
- Shandong Technological Center of Oceanographic Instrumentation Co., Ltd 37 Miaoling Road Qingdao 266061 P. R. China
| | - Huiling Li
- Innovation and Development Institute of Shangdong Province Jinan 250101 P. R. China
| | - Zike Jiang
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Marine Monitoring Instrument Equipment Technology, National Engineering and Technological Research Center of Marine Monitoring Equipment 37 Miaoling Road Qingdao 266061 P. R. China
| |
Collapse
|
9
|
Zhang C, Wang L, Xu Y, Huang Y, Huang J, Zhu J, Wang W, Li W, Sun A, Li X, Zhang H, Li J. Discovery of novel dual RAGE/SERT inhibitors for the potential treatment of the comorbidity of Alzheimer's disease and depression. Eur J Med Chem 2022; 236:114347. [DOI: 10.1016/j.ejmech.2022.114347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/08/2023]
|
10
|
Singh K, Bhatia R, Kumar B, Singh G, Monga V. Design Strategies, Chemistry and Therapeutic Insights of Multi-target Directed Ligands as Antidepressant Agents. Curr Neuropharmacol 2022; 20:1329-1358. [PMID: 34727859 PMCID: PMC9881079 DOI: 10.2174/1570159x19666211102154311] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/21/2021] [Accepted: 10/19/2021] [Indexed: 11/22/2022] Open
Abstract
Depression is one of the major disorders of the central nervous system worldwide and causes disability and functional impairment. According to the World Health Organization, around 265 million people worldwide are affected by depression. Currently marketed antidepressant drugs take weeks or even months to show anticipated clinical efficacy but remain ineffective in treating suicidal thoughts and cognitive impairment. Due to the multifactorial complexity of the disease, single-target drugs do not always produce satisfactory results and lack the desired level of therapeutic efficacy. Recent literature reports have revealed improved therapeutic potential of multi-target directed ligands due to their synergistic potency and better safety. Medicinal chemists have gone to great extents to design multitarget ligands by generating structural hybrids of different key pharmacophores with improved binding affinities and potency towards different receptors or enzymes. This article has compiled the design strategies of recently published multi-target directed ligands as antidepressant agents. Their biological evaluation, structural-activity relationships, mechanistic and in silico studies have also been described. This article will prove to be highly useful for the researchers to design and develop multi-target ligands as antidepressants with high potency and therapeutic efficacy.
Collapse
Affiliation(s)
- Karanvir Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Rohit Bhatia
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Vikramdeep Monga
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda-151401, Punjab, India
| |
Collapse
|
11
|
Li X, Li J, Huang Y, Gong Q, Fu Y, Xu Y, Huang J, You H, Zhang D, Zhang D, Mao F, Zhu J, Wang H, Zhang H, Li J. The novel therapeutic strategy of vilazodone-donepezil chimeras as potent triple-target ligands for the potential treatment of Alzheimer's disease with comorbid depression. Eur J Med Chem 2021; 229:114045. [PMID: 34922191 DOI: 10.1016/j.ejmech.2021.114045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/24/2021] [Accepted: 12/05/2021] [Indexed: 12/14/2022]
Abstract
Depression is one of the most frequent comorbid psychiatric symptoms of Alzheimer's disease (AD), and no efficacious drugs have been approved specifically for this purpose thus far. Herein, we proposed a novel therapeutic strategy that merged the key pharmacophores of the antidepressant vilazodone (5-HT1A receptor partial agonist and serotonin transporter inhibitor) and the anti-AD drug donepezil (acetylcholinesterase inhibitor) together to develop a series of multi-target-directed ligands for potential therapy of the comorbidity of AD and depression. Accordingly, 55 vilazodone-donepezil chimeric derivatives were designed and synthesized, and their triple-target activities against acetylcholinesterase, 5-HT1A receptor, and serotonin transporter were systematically evaluated. Among them, compound 5 displayed strong triple-target bioactivities in vitro, low hERG potassium channel inhibition and acceptable brain distribution. Importantly, oral intake of 5 mg/kg of the compound 5 dihydrochloride significantly alleviated the depressive symptoms and ameliorated cognitive dysfunction in mouse models. In brief, these results highlight vilazodone-donepezil chimeras as a prospective therapeutic approach for the treatment of the comorbidity of AD and depression.
Collapse
Affiliation(s)
- Xiaokang Li
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Jinwen Li
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Yunyuan Huang
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Qi Gong
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Yan Fu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Yixiang Xu
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Junyang Huang
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Haolan You
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Dong Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Dan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Fei Mao
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Jin Zhu
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Huan Wang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
| | - Haiyan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China; Yunnan Key Laboratory of Screening and Research on Anti-pathogenic Plant Resources from West Yunnan, College of Pharmacy, Dali University, 5 Xue Ren Road, Dali, Yunnan, 671000, China; Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
12
|
Choudhury S, Jena S, Sahoo DK, Shekh S, Kar RK, Dhakad A, Gowd KH, Biswal HS. Gram-Scale Synthesis of 1,8-Naphthyridines in Water: The Friedlander Reaction Revisited. ACS OMEGA 2021; 6:19304-19313. [PMID: 34337267 PMCID: PMC8320145 DOI: 10.1021/acsomega.1c02798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
The products of the Friedlander reaction, i.e., 1,8-naphthyridines, have far-reaching impacts in materials science, chemical biology, and medicine. The reported synthetic methodologies elegantly orchestrate the diverse synthetic routes of naphthyridines but require harsh reaction conditions, organic solvents, and expensive metal catalysts. Here, we introduce gram-scale synthesis of 1,8-naphthyridines in water using an inexpensive and biocompatible ionic liquid (IL) as a catalyst. This is the first-ever report on the synthesis of naphthyridines in water. This is a one-step reaction, and the product separation is relatively easy. The choline hydroxide (ChOH) is used as a metal-free, nontoxic, and water-soluble catalyst. In comparison to other catalysts reported in the literature, ChOH has the advantage of forming an additional hydrogen bond with the reactants, which is the vital step for the reaction to happen in water. Density functional theory (DFT) and noncovalent interaction (NCI) plot index analysis provide the plausible reaction mechanism for the catalytic cycle and confirm that hydrogen bonds with the IL catalyst are pivotal to facilitate the reaction. Molecular docking and molecular dynamics (MD) simulations are also performed to demonstrate the potentialities of the newly synthesized products as drugs. Through MD simulations, it was established that the tetrahydropyrido derivative of naphthyridine (10j) binds to the active sites of the ts3 human serotonin transporter (hSERT) (PDB ID: 6AWO) without perturbing the secondary structure, suggesting that 10j can be a potential preclinical drug candidate for hSERT inhibition and depression treatment.
Collapse
Affiliation(s)
- Shubhranshu
Shekhar Choudhury
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Subhrakant Jena
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Dipak Kumar Sahoo
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Shamasoddin Shekh
- Department
of Chemistry, Central University of Karnataka, Kalaburagi 585367, Karnataka, India
| | - Rajiv K. Kar
- Fritz
Haber Center for Molecular Dynamics, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Ambuj Dhakad
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Konkallu Hanumae Gowd
- Department
of Chemistry, Central University of Karnataka, Kalaburagi 585367, Karnataka, India
| | - Himansu S. Biswal
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| |
Collapse
|
13
|
Szałaj N, Godyń J, Jończyk J, Pasieka A, Panek D, Wichur T, Więckowski K, Zaręba P, Bajda M, Pislar A, Malawska B, Sabate R, Więckowska A. Multidirectional in vitro and in cellulo studies as a tool for identification of multi-target-directed ligands aiming at symptoms and causes of Alzheimer's disease. J Enzyme Inhib Med Chem 2021; 35:1944-1952. [PMID: 33092411 PMCID: PMC7594877 DOI: 10.1080/14756366.2020.1835882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Effective therapy of Alzheimer's disease (AD) requires treatment with a combination of drugs that modulate various pathomechanisms contributing to the disease. In our research, we have focused on the development of multi-target-directed ligands - 5-HT6 receptor antagonists and cholinesterase inhibitors - with disease-modifying properties. We have performed extended in vitro (FRET assay) and in cellulo (Escherichia coli model of protein aggregation) studies on their β-secretase, tau, and amyloid β aggregation inhibitory activity. Within these multifunctional ligands, we have identified compound 17 with inhibitory potency against tau and amyloid β aggregation in in cellulo assay of 59% and 56% at 10 µM, respectively, hBACE IC50=4 µM, h5TH6 K i=94 nM, hAChE IC50=26 nM, and eqBuChE IC50=5 nM. This study led to the development of multifunctional ligands with a broad range of biological activities crucial not only for the symptomatic but also for the disease-modifying treatment of AD.
Collapse
Affiliation(s)
- Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Jakub Jończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Anna Pasieka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz Wichur
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Krzysztof Więckowski
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Paula Zaręba
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Marek Bajda
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Anja Pislar
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Raimon Sabate
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
14
|
Kumar B, Thakur A, Dwivedi AR, Kumar R, Kumar V. Multi-Target-Directed Ligands as an Effective Strategy for the Treatment of Alzheimer's Disease. Curr Med Chem 2021; 29:1757-1803. [PMID: 33982650 DOI: 10.2174/0929867328666210512005508] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder, and multiple pathological factors are believed to be involved in the genesis and progression of the disease. A number of hypotheses, including Acetylcholinesterase, Monoamine oxidase, β-Amyloid, Tau protein, etc., have been proposed for the initiation and progression of the disease. At present, acetylcholine esterase inhibitors and memantine (NMDAR antagonist) are the only approved therapies for the symptomatic management of AD. Most of these single-target drugs have miserably failed in the treatment or halting the progression of the disease. Multi-factorial diseases like AD require complex treatment strategies that involve simultaneous modulation of a network of interacting targets. Since the last few years, Multi-Target-Directed Ligands (MTDLs) strategy, drugs that can simultaneously hit multiple targets, is being explored as an effective therapeutic approach for the treatment of AD. In the current review article, the authors have briefly described various pathogenic pathways associated with AD. The importance of Multi-Target-Directed Ligands and their design strategies in recently reported articles have been discussed in detail. Potent leads are identified through various structure-activity relationship studies, and their drug-like characteristics are described. Recently developed promising compounds have been summarized in the article. Some of these MTDLs with balanced activity profiles against different targets have the potential to be developed as drug candidates for the treatment of AD.
Collapse
Affiliation(s)
- Bhupinder Kumar
- Central University of Punjab Department of Pharmaceutical Sciences and Natural Products, India
| | - Amandeep Thakur
- Central University of Punjab Department of Pharmaceutical Sciences and Natural Products, India
| | | | - Rakesh Kumar
- Central University of Punjab, Bathinda, Punjab-151001, India
| | - Vinod Kumar
- Department of Chemistry, Central University of Punjab, Bathinda, Punjab-151001, India
| |
Collapse
|
15
|
Design and synthesis of novel tacrine-indole hybrids as potential multitarget-directed ligands for the treatment of Alzheimer's disease. Future Med Chem 2021; 13:785-804. [PMID: 33829876 DOI: 10.4155/fmc-2020-0184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The authors report on the synthesis and biological evaluation of new compounds whose structure combines tacrine and indole moieties. Tacrine-indole heterodimers were designed to inhibit cholinesterases and β-amyloid formation, and to cross the blood-brain barrier. The most potent new acetylcholinesterase inhibitors were compounds 3c and 4d (IC50 = 25 and 39 nM, respectively). Compound 3c displayed considerably higher selectivity for acetylcholinesterase relative to human plasma butyrylcholinesterase in comparison to compound 4d (selectivity index: IC50 [butyrylcholinesterase]/IC50 [acetylcholinesterase] = 3 and 0.6, respectively). Furthermore, compound 3c inhibited β-amyloid-dependent amyloid nucleation in the yeast-based prion nucleation assay and displayed no dsDNA destabilizing interactions with DNA. Compounds 3c and 4d displayed a high probability of crossing the blood-brain barrier. The results support the potential of 3c for future development as a dual-acting therapeutic agent in the prevention and/or treatment of Alzheimer's disease.
Collapse
|
16
|
Manzoor S, Prajapati SK, Majumdar S, Raza MK, Gabr MT, Kumar S, Pal K, Rashid H, Kumar S, Krishnamurthy S, Hoda N. Discovery of new phenyl sulfonyl-pyrimidine carboxylate derivatives as the potential multi-target drugs with effective anti-Alzheimer's action: Design, synthesis, crystal structure and in-vitro biological evaluation. Eur J Med Chem 2021; 215:113224. [PMID: 33582578 DOI: 10.1016/j.ejmech.2021.113224] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is multifactorial, progressive neurodegeneration with impaired behavioural and cognitive functions. The multitarget-directed ligand (MTDL) strategies are promising paradigm in drug development, potentially leading to new possible therapy options for complex AD. Herein, a series of novel MTDLs phenylsulfonyl-pyrimidine carboxylate (BS-1 to BS-24) derivatives were designed and synthesized for AD treatment. All the synthesized compounds were validated by 1HNMR, 13CNMR, HRMS, and BS-19 were structurally validated by X-Ray single diffraction analysis. To evaluate the plausible binding affinity of designed compounds, molecular docking study was performed, and the result revealed their significant interaction with active sites of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). The synthesized compounds displayed moderate to excellent in vitro enzyme inhibitory activity against AChE and BuChE at nanomolar (nM) concentration. Among 24 compounds (BS-1 to BS-24), the optimal compounds (BS-10 and BS-22) displayed potential inhibition against AChE; IC50 = 47.33 ± 0.02 nM and 51.36 ± 0.04 nM and moderate inhibition against BuChE; IC50 = 159.43 ± 0.72 nM and 153.3 ± 0.74 nM respectively. In the enzyme kinetics study, the compound BS-10 displayed non-competitive inhibition of AChE with Ki = 8 nM. Respective compounds BS-10 and BS-22 inhibited AChE-induced Aβ1-42 aggregation in thioflavin T-assay at 10 μM and 20 μM, but BS-10 at 10 μM and 20 μM concentrations are found more potent than BS-22. In addition, the aggregation properties were determined by the dynamic light scattering (DLS) and was found that BS-10 and BS-22 could significantly inhibit self-induced as well as AChE-induced Aβ1-42 aggregation. The effect of compounds (BS-10 and BS-22) on the viability of MC65 neuroblastoma cells and their capability to cross the blood-brain barrier (BBB) in PAMPA-BBB were further studied. Further, in silico approach was applied to analyze physicochemical and pharmacokinetics properties of the designed compounds via the SwissADME and PreADMET server. Hence, the novel phenylsulfonyl-pyrimidine carboxylate derivatives can act as promising leads in the development of AChE inhibitors and Aβ disaggregator for the treatment of AD.
Collapse
Affiliation(s)
- Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Santosh Kumar Prajapati
- Neurotherapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P, 221005, India
| | - Shreyasi Majumdar
- Neurotherapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P, 221005, India
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Moustafa T Gabr
- Department of Radiology, Stanford University, Stanford, CA, 94305, United States
| | - Shivani Kumar
- University School of Biotechnology Guru Gobind Singh Indraprastha University Dwarka, Sector 16C, New Delhi, 110078, India
| | - Kavita Pal
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Haroon Rashid
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Suresh Kumar
- University School of Biotechnology Guru Gobind Singh Indraprastha University Dwarka, Sector 16C, New Delhi, 110078, India
| | - Sairam Krishnamurthy
- Neurotherapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P, 221005, India.
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
17
|
Bowroju SK, Mainali N, Ayyadevara S, Penthala NR, Krishnamachari S, Kakraba S, Reis RJS, Crooks PA. Design and Synthesis of Novel Hybrid 8-Hydroxy Quinoline-Indole Derivatives as Inhibitors of Aβ Self-Aggregation and Metal Chelation-Induced Aβ Aggregation. Molecules 2020; 25:E3610. [PMID: 32784464 PMCID: PMC7463714 DOI: 10.3390/molecules25163610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 02/03/2023] Open
Abstract
A series of novel hybrid 8-hydroxyquinoline-indole derivatives (7a-7e, 12a-12b and 18a-18h) were synthesized and screened for inhibitory activity against self-induced and metal-ion induced Aβ1-42 aggregation as potential treatments for Alzheimer's disease (AD). In vitro studies identified the most inhibitory compounds against self-induced Aβ1-42 aggregation as 18c, 18d and 18f (EC50 = 1.72, 1.48 and 1.08 µM, respectively) compared to the known anti-amyloid drug, clioquinol (1, EC50 = 9.95 µM). The fluorescence of thioflavin T-stained amyloid formed by Aβ1-42 aggregation in the presence of Cu2+ or Zn2+ ions was also dramatically decreased by treatment with 18c, 18d and 18f. The most potent hybrid compound 18f afforded 82.3% and 88.3% inhibition, respectively, against Cu2+- induced and Zn2+- induced Aβ1-42 aggregation. Compounds 18c, 18d and 18f were shown to be effective in reducing protein aggregation in HEK-tau and SY5Y-APPSw cells. Molecular docking studies with the most active compounds performed against Aβ1-42 peptide indicated that the potent inhibitory activity of 18d and 18f were predicted to be due to hydrogen bonding interactions, π-π stacking interactions and π-cation interactions with Aβ1-42, which may inhibit both self-aggregation as well as metal ion binding to Aβ1-42 to favor the inhibition of Aβ1-42 aggregation.
Collapse
Affiliation(s)
- Suresh K. Bowroju
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.K.B.); (N.R.P.)
| | - Nirjal Mainali
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (S.K.)
| | - Srinivas Ayyadevara
- Central Arkansas Veterans Healthcare Service, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.K.); (S.A.)
| | - Narsimha R. Penthala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.K.B.); (N.R.P.)
| | - Sesha Krishnamachari
- Central Arkansas Veterans Healthcare Service, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.K.); (S.A.)
| | - Samuel Kakraba
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (S.K.)
| | - Robert J. Shmookler Reis
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (S.K.)
- Central Arkansas Veterans Healthcare Service, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.K.); (S.A.)
- Department of Geriatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Peter A. Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.K.B.); (N.R.P.)
| |
Collapse
|
18
|
Li X, Lu J, Xu Y, Wang J, Qiu X, Fan L, Li B, Liu W, Mao F, Zhu J, Shen X, Li J. Discovery of nitazoxanide-based derivatives as autophagy activators for the treatment of Alzheimer's disease. Acta Pharm Sin B 2020; 10:646-666. [PMID: 32322468 PMCID: PMC7161708 DOI: 10.1016/j.apsb.2019.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/03/2019] [Accepted: 07/17/2019] [Indexed: 12/26/2022] Open
Abstract
Drug repurposing is an efficient strategy for new drug discovery. Our latest study found that nitazoxanide (NTZ), an approved anti-parasite drug, was an autophagy activator and could alleviate the symptom of Alzheimer's disease (AD). In order to further improve the efficacy and discover new chemical entities, a series of NTZ-based derivatives were designed, synthesized, and evaluated as autophagy activator against AD. All compounds were screened by the inhibition of phosphorylation of p70S6K, which was the direct substrate of mammalian target of rapamycin (mTOR) and its phosphorylation level could reflect the mTOR-dependent autophagy level. Among these analogs, compound 22 exhibited excellent potency in promoting β-amyloid (Aβ) clearance, inhibiting tau phosphorylation, as well as stimulating autophagy both in vitro and in vivo. What's more, 22 could effectively improve the memory and cognitive impairments in APP/PS1 transgenic AD model mice. These results demonstrated that 22 was a potential candidate for the treatment of AD.
Collapse
Key Words
- AChEIs, acetylcholinesterase inhibitors
- AD, Alzheimer's disease
- APP, amyloid precursor protein
- Alzheimer's disease
- Autophagy
- Aβ, β-amyloid
- BBB, blood–brain barrier
- CNS, central nervous system
- MWM, Morris Water Maze
- NCEs, new chemical entities
- NFTs, neurofibrillary tangles
- NMDA, N-methyl-d-aspartate
- NTZ, nitazoxanide
- Nitazoxanide
- PAMPA, parallel artificial membrane permeation assay
- PBL, porcine brain lipid
- SPs, senile plaques
- Tau protein
- WORT, wortmannin
- mTOR, mammalian target of rapamycin
- β-amyloid
Collapse
|
19
|
Zhang Y, Zheng G, Fu T, Hong J, Li F, Yao X, Xue W, Zhu F. The binding mode of vilazodone in the human serotonin transporter elucidated by ligand docking and molecular dynamics simulations. Phys Chem Chem Phys 2020; 22:5132-5144. [PMID: 32073004 DOI: 10.1039/c9cp05764a] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vilazodone is a novel antidepressant used for the treatment of major depressive disorder (MDD) with a primary action mechanism of inhibiting the human serotonin reuptake transporter (hSERT) and acting as a 5-HT1A receptor partial agonist. The interaction between vilazodone and the 5-HT1A receptor has been reported, however, the binding mode of vilazodone in the hSERT remains elusive. In the current study, to elucidate the molecular mechanism of vilazodone binding in the hSERT, the drug and its five analogs were docked into the hSERT crystal structure as initial conformations and were sampled by 400 ns molecular dynamics (MD) simulations. Through the analysis of the profiles of protein-ligand binding free energies, interaction fingerprints, and conformational rearrangements, the binding mode of vilazodone in the hSERT was revealed. As a result, unlike the classical antidepressants located in the S1 site of the hSERT, vilazodone adopted a linear pose in the binding pocket. Its arylpiperazine fragment occupies the central site (S1) and interacts with Y95, D98, I172, Y176, F335, F341, S438, and T439, while the indole fragment extends to the allosteric site (S2) via interacting with the ionic switch (R104/E403) between the two sites. The new insights obtained are not only helpful in understanding the binding mode of vilazodone in the hSERT, but also provide valuable guidance to the discovery of novel antidepressant drugs.
Collapse
Affiliation(s)
- Yang Zhang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Przybyłowska M, Kowalski S, Dzierzbicka K, Inkielewicz-Stepniak I. Therapeutic Potential of Multifunctional Tacrine Analogues. Curr Neuropharmacol 2019; 17:472-490. [PMID: 29651948 PMCID: PMC6520589 DOI: 10.2174/1570159x16666180412091908] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/25/2018] [Accepted: 04/04/2018] [Indexed: 12/12/2022] Open
Abstract
Abstract: Tacrine is a potent inhibitor of cholinesterases (acetylcholinesterase and butyrylcholinesterase) that shows limiting clinical application by liver toxicity. In spite of this, analogues of tacrine are considered as a model inhibitor of cholinesterases in the therapy of Alzheimer’s disease. The interest in these compounds is mainly related to a high variety of their structure and biological properties. In the present review, we have described the role of cholinergic transmission and treatment strategies in Alzheimer’s disease as well as the synthesis and biological activity of several recently developed classes of multifunctional tacrine analogues and hybrids, which consist of a new paradigm to treat Alzheimer’s disease. We have also reported potential of these analogues in the treatment of Alzheimer’s diseases in various experimental systems.
Collapse
Affiliation(s)
- Maja Przybyłowska
- Department of Organic Chemistry, Gdansk University of Technology, 11/12 G. Narutowicza Street, 80-233, Gdansk, Poland
| | - Szymon Kowalski
- Department of Medical Chemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry, Gdansk University of Technology, 11/12 G. Narutowicza Street, 80-233, Gdansk, Poland
| | | |
Collapse
|
21
|
Pan T, Xie S, Zhou Y, Hu J, Luo H, Li X, Huang L. Dual functional cholinesterase and PDE4D inhibitors for the treatment of Alzheimer’s disease: Design, synthesis and evaluation of tacrine-pyrazolo[3,4-b]pyridine hybrids. Bioorg Med Chem Lett 2019; 29:2150-2152. [DOI: 10.1016/j.bmcl.2019.06.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/21/2019] [Accepted: 06/28/2019] [Indexed: 10/26/2022]
|
22
|
Rodríguez-Soacha DA, Scheiner M, Decker M. Multi-target-directed-ligands acting as enzyme inhibitors and receptor ligands. Eur J Med Chem 2019; 180:690-706. [PMID: 31401465 DOI: 10.1016/j.ejmech.2019.07.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
Abstract
In this review, we present the latest advances in the field of multi-target-directed ligand (MTDL) design for the treatment of various complex pathologies of multifactorial origin. In particular, latest findings in the field of MTDL design targeting both an enzyme and a receptor are presented for different diseases such as Alzheimer's disease (AD), depression, addiction, glaucoma, non-alcoholic steatohepatitis and pain and inflammation. The ethology of the diseases is briefly described, with special emphasis on how the MTDL can evolve into novel therapies that replace the classic pharmacological dogma "one target one disease". Considering the current needs for therapy adherence improvement, it is exposed as from the medicinal chemistry, different molecular scaffolds are studied. With the use of structure activity relationship studies and molecular optimization, new hybrid molecules are generated with improved biological properties acting at two biologically very distinct targets.
Collapse
Affiliation(s)
- Diego Alejandro Rodríguez-Soacha
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany.
| |
Collapse
|
23
|
Du H, Liu X, Xie J, Ma F. Novel Deoxyvasicinone-Donepezil Hybrids as Potential Multitarget Drug Candidates for Alzheimer's Disease. ACS Chem Neurosci 2019; 10:2397-2407. [PMID: 30720268 DOI: 10.1021/acschemneuro.8b00699] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In this study, we designed and synthesized a series of deoxyvasicinone-donepezil hybrids and determined whether they could be used as novel multitarget inhibitors for Alzheimer's disease. In vitro studies showed that most of the hybrids demonstrated moderate to potent inhibition of hAChE, BACE1, and Aβ1-42 aggregation. In particular, the hybrids 10a, 10d, 11a, and 11j exhibited excellent inhibitory activities against hAChE (IC50 = 56.14, 5.91, 3.29, and 8.65 nM, respectively), BACE1 (IC50 = 0.834, 0.167, 0.129, and 0.085 μM, respectively), and Aβ1-42 aggregation (IC50 = 13.26, 19.43, 9.26, and 5.41 μM, respectively). In addition, 10a and 11a exhibited very low cytotoxicity and showed remarkable neuroprotective activity against Aβ1-42-induced damage in SH-SY5Y cells.
Collapse
Affiliation(s)
- Hongtao Du
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
- College of Plant Protection, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Xinlian Liu
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
- Tea Plant Biology Key Laboratory of Henan Province, Xinyang 464000, China
| | - Jusen Xie
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
- Tea Plant Biology Key Laboratory of Henan Province, Xinyang 464000, China
| | - Fang Ma
- School of Geographic Sciences, Xinyang Normal University, Xinyang 464000, China
| |
Collapse
|
24
|
Mahdavi M, Hariri R, Mirfazli SS, Lotfian H, Rastergari A, Firuzi O, Edraki N, Larijani B, Akbarzadeh T, Saeedi M. Synthesis and Biological Activity of Some Benzochromenoquinolinones: Tacrine Analogs as Potent Anti-Alzheimer's Agents. Chem Biodivers 2019; 16:e1800488. [PMID: 30720917 DOI: 10.1002/cbdv.201800488] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/04/2019] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is a well-known neurodegenerative disorder affecting millions of old people worldwide and the corresponding epidemiological data emphasize the importance of the disease. As AD is a multifactorial illness, various single target directed drugs that have reached clinical trials have failed. Therefore, various factors associated with outset of AD have been considered in targeted drug discovery. In this work, various benzochromenoquinolinones were synthesized and evaluated for their cholinesterase and BACE1 inhibitory activities as well as neuroprotective and metal-chelating properties. Among the synthesized compounds, 14-amino-13-(3-nitrophenyl)-2,3,4,13-tetrahydro-1H-benzo[6,7]chromeno[2,3-b]quinoline-7,12-dione (6m) depicted the best inhibitory activity toward acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) with IC50 s of 0.86 and 6.03 μm, respectively. Also, the compound could inhibit β-secretase 1 (BACE1) with IC50 =19.60 μm and showed metal chelating ability toward Cu2+ , Fe2+ , and Zn2+ . In addition, docking study demonstrated desirable interactions of compound 6m with amino acid residues characterizing AChE, BChE, and BACE1.
Collapse
Affiliation(s)
- Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Roshanak Hariri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran, University of Medical Sciences, Tehran, 1416753955, Iran
| | - Seyedeh Sara Mirfazli
- Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Hania Lotfian
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran, University of Medical Sciences, Tehran, 1416753955, Iran
| | - Arezoo Rastergari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran, University of Medical Sciences, Tehran, 1416753955, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, 71345-1978, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, 71345-1978, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran, University of Medical Sciences, Tehran, 1416753955, Iran.,Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Mina Saeedi
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, 1416753955, Iran.,Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| |
Collapse
|
25
|
Barré A, Azzouz R, Gembus V, Papamicaël C, Levacher V. Design, Synthesis, and In Vitro Biological Activities of a Bio-Oxidizable Prodrug to Deliver Both ChEs and DYRK1A Inhibitors for AD Therapy. Molecules 2019; 24:E1264. [PMID: 30939771 PMCID: PMC6479981 DOI: 10.3390/molecules24071264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 01/28/2023] Open
Abstract
Despite their side effects, cholinesterase (ChE) inhibitors remain the only approved drugs to treat Alzheimer's disease patients, along with the N-methyl-d-aspartate (NMDA) receptor antagonist memantine. In the last few years, the dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) has also been studied as a promising target for the development of new drugs for this pathology. In this context, and based on our previous characterization of bio-oxidizable prodrugs of potent acetylcholinesterase (AChE) inhibitors, we envisioned a strategy involving the synthesis of a bio-oxidizable prodrug of both ChE and DYRK1A inhibitors. To this end, we fixed our interest on a known potent inhibitor of DYRK1A, namely INDY. The designed prodrug of both ChE and DYRK1A inhibitors was successfully synthesized, connecting both inhibitors by a carbonate link. This prodrug and its corresponding drug were then evaluated as ChEs and DYRK1A inhibitors. Remarkably, in vitro results were in accordance with the starting hypothesis, showing a relative inactivity of the prodrug against DYRK1A and ChEs and a potent inhibition of ChEs by the oxidized form. Molecular docking and kinetic studies of ChE inhibition by the active compound are also discussed in this report.
Collapse
Affiliation(s)
- Anaïs Barré
- VFP Therapies R&D; 1 rue Tesnière, 76130 Mont Saint-Aignan, France.
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France.
| | - Rabah Azzouz
- VFP Therapies R&D; 1 rue Tesnière, 76130 Mont Saint-Aignan, France.
| | - Vincent Gembus
- VFP Therapies R&D; 1 rue Tesnière, 76130 Mont Saint-Aignan, France.
| | - Cyril Papamicaël
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France.
| | - Vincent Levacher
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France.
| |
Collapse
|
26
|
Mishra P, Kumar A, Panda G. Anti-cholinesterase hybrids as multi-target-directed ligands against Alzheimer’s disease (1998–2018). Bioorg Med Chem 2019; 27:895-930. [DOI: 10.1016/j.bmc.2019.01.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/15/2019] [Accepted: 01/23/2019] [Indexed: 01/09/2023]
|
27
|
de los Ríos C, Marco-Contelles J. Tacrines for Alzheimer's disease therapy. III. The PyridoTacrines. Eur J Med Chem 2019; 166:381-389. [DOI: 10.1016/j.ejmech.2019.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/25/2019] [Accepted: 02/02/2019] [Indexed: 11/26/2022]
|
28
|
Wang H, Zhang H. Reconsideration of Anticholinesterase Therapeutic Strategies against Alzheimer's Disease. ACS Chem Neurosci 2019; 10:852-862. [PMID: 30521323 DOI: 10.1021/acschemneuro.8b00391] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is well-known as a severe neurodegeneration disease involving complicated etiologies, and cholinesterase inhibition remain the prevailing mode of clinical intervention in AD management. Although most clinically applied cholinesterase inhibitors (ChEIs) achieve limited clinical outcomes, research on the central cholinergic system is still thriving. Recently, an impressive amount of knowledge regarding novel acetylcholinesterase functions, as well as the close association between the central cholinergic system and other key elements for AD pathogenesis, has accumulated, highlighting that this field still has great potential for future drug development. In contrast to the overwhelmingly disappointing clinical therapeutic effects of various disease-modifying drug candidates, interesting evidence has continued to emerge over the past 20 years from the wealth of preclinical and clinical data on the usage of ChEIs, indicating underestimated clinical benefits due to physician ambivalence, a lack of persistent treatment, and inappropriate medication times or doses. Here we pinpoint several topics fit for future attention, focusing on the updated cholinergic hypothesis, especially the pleiotropic relationships with key pathogenetic signaling pathways and functions in AD, as well as possible novel therapeutic strategies, including novel ChEIs and cholinesterase inhibition-based innovative multifunctional therapeutic candidates. We intend to strengthen the future value of the precise application of cholinergic drugs, especially novel ChEIs, as a cornerstone pharmacological approach to AD treatment, either alone or in combination with other targets, to relieve symptoms and to modify disease progression.
Collapse
Affiliation(s)
- Huan Wang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Haiyan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| |
Collapse
|
29
|
Synthesis and evaluation of clioquinol-rolipram/roflumilast hybrids as multitarget-directed ligands for the treatment of Alzheimer's disease. Eur J Med Chem 2019; 163:512-526. [DOI: 10.1016/j.ejmech.2018.12.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023]
|
30
|
Xu Y, Zhang J, Wang H, Mao F, Bao K, Liu W, Zhu J, Li X, Zhang H, Li J. Rational Design of Novel Selective Dual-Target Inhibitors of Acetylcholinesterase and Monoamine Oxidase B as Potential Anti-Alzheimer's Disease Agents. ACS Chem Neurosci 2019; 10:482-496. [PMID: 30110536 DOI: 10.1021/acschemneuro.8b00357] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multifunctional agents aiming at cholinesterases (ChEs) and monoamine oxidases (MAOs) are promising therapy for Alzheimer's disease (AD). Herein, a series of novel propargylamine-modified pyrimidinylthiourea derivatives (1-4) were designed and synthesized as dual inhibitors of ChEs and MAOs with other functions against AD. Most of these derivatives inhibited ChEs and MAOs with IC50 values in the micro- or nanomolar ranges. Compound 1c displayed the dual functional profile of targeting the AChE (IC50 = 0.032 ± 0.007 μM) and MAO-B (IC50 = 2.117 ± 0.061 μM), along with the improved blood-brain barrier (BBB) permeability, antioxidant ability, and good copper chelating property in vitro. Animal studies showed that compound 1c·HCl could inhibit the cerebral AChE/MAO-B activities and alleviate scopolamine-induced cognitive impairment in mice. Combined with good oral bioavailability ( F = 45.55%), these findings demonstrated that compound 1c may be a potent brain permeable multifunctional candidate for the treatment of AD.
Collapse
Affiliation(s)
- Yixiang Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jian Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Huan Wang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Fei Mao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Keting Bao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Wenwen Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jin Zhu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Xiaokang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Haiyan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| |
Collapse
|
31
|
Akkachairin B, Tummatorn J, Khamsuwan N, Thongsornkleeb C, Ruchirawat S. Domino N2-Extrusion–Cyclization of Alkynylarylketone Derivatives for the Synthesis of Indoloquinolines and Carbocycle-Fused Quinolines. J Org Chem 2018; 83:11254-11268. [DOI: 10.1021/acs.joc.8b01851] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Bhornrawin Akkachairin
- Program on Chemical Biology, Chulabhorn Graduate Institute, Center of Excellence on Environmental Health and Toxicology (EHT), Ministry of Education, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
| | - Jumreang Tummatorn
- Program on Chemical Biology, Chulabhorn Graduate Institute, Center of Excellence on Environmental Health and Toxicology (EHT), Ministry of Education, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
| | - Narumol Khamsuwan
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
| | - Charnsak Thongsornkleeb
- Program on Chemical Biology, Chulabhorn Graduate Institute, Center of Excellence on Environmental Health and Toxicology (EHT), Ministry of Education, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
| | - Somsak Ruchirawat
- Program on Chemical Biology, Chulabhorn Graduate Institute, Center of Excellence on Environmental Health and Toxicology (EHT), Ministry of Education, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Laksi, Bangkok 10210, Thailand
| |
Collapse
|
32
|
Liu W, Wang H, Li X, Xu Y, Zhang J, Wang W, Gong Q, Qiu X, Zhu J, Mao F, Zhang H, Li J. Design, synthesis and evaluation of vilazodone-tacrine hybrids as multitarget-directed ligands against depression with cognitive impairment. Bioorg Med Chem 2018; 26:3117-3125. [PMID: 29729987 DOI: 10.1016/j.bmc.2018.04.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/08/2018] [Accepted: 04/16/2018] [Indexed: 01/21/2023]
Abstract
Depression, a severe mental disease, is greatly difficult to treat and easy to induce other neuropsychiatric symptoms, the most frequent one is cognitive impairment. In this study, a series of novel vilazodone-tacrine hybrids were designed, synthesized and evaluated as multitarget agents against depression with cognitive impairment. Most compounds exhibited good multitarget activities and appropriate blood-brain barrier permeability. Specifically, compounds 1d and 2a exhibited excellent 5-HT1A agonist activities (1d, EC50 = 0.36 ± 0.08 nM; 2a, EC50 = 0.58 ± 0.14 nM) and 5-HT reuptake inhibitory activities (1d, IC50 = 20.42 ± 6.60 nM; 2a, IC50 = 22.10 ± 5.80 nM). In addition, they showed moderate ChE inhibitory activities (1d, AChE IC50 = 1.72 ± 0.217 μM, BuChE IC50 = 0.34 ± 0.03 μM; 2a, AChE IC50 = 2.36 ± 0.34 μM, BuChE IC50 = 0.10 ± 0.01 μM). Good multitarget activities with goodt blood-brain barrier permeability of 1d and 2a make them good lead compounds for the further study of depression with cognitive impairment.
Collapse
Affiliation(s)
- Wenwen Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Huan Wang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaokang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Yixiang Xu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jian Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Wei Wang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Qi Gong
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Xiaoxia Qiu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jin Zhu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Fei Mao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China.
| | - Haiyan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Jian Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China.
| |
Collapse
|