Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Discovery, Biological Evaluation, and Crystal Structure of a Novel Nanomolar Selective Butyrylcholinesterase Inhibitor
My Activity
    Article

    Discovery, Biological Evaluation, and Crystal Structure of a Novel Nanomolar Selective Butyrylcholinesterase Inhibitor
    Click to copy article linkArticle link copied!

    View Author Information
    Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
    IBS, University Grenoble Alpes, F-38044 Grenoble, France
    § IBS, CNRS, F-38044 Grenoble, France
    IBS, CEA, F-38044 Grenoble, France
    Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
    # Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
    *For S.G.: phone, +386-1-4769500; fax, +386-1-4258031; E-mail, [email protected]
    *For J.P.C.: phone, +33457428515; fax, +33476501890; E-mail, [email protected]
    Other Access OptionsSupporting Information (1)

    Journal of Medicinal Chemistry

    Cite this: J. Med. Chem. 2014, 57, 19, 8167–8179
    Click to copy citationCitation copied!
    https://doi.org/10.1021/jm501195e
    Published September 16, 2014
    Copyright © 2014 American Chemical Society

    Abstract

    Click to copy section linkSection link copied!
    Abstract Image

    Butyrylcholinesterase (BChE) is regarded as a promising drug target as its levels and activity significantly increase in the late stages of Alzheimer’s disease. To discover novel BChE inhibitors, we used a hierarchical virtual screening protocol followed by biochemical evaluation of 40 highest scoring hit compounds. Three of the compounds identified showed significant inhibitory activities against BChE. The most potent, compound 1 (IC50 = 21.3 nM), was resynthesized and resolved into its pure enantiomers. A high degree of stereoselective activity was revealed, and a dissociation constant of 2.7 nM was determined for the most potent stereoisomer (+)-1. The crystal structure of human BChE in complex with compound (+)-1 was solved, revealing the binding mode and providing clues for potential optimization. Additionally, compound 1 inhibited amyloid β1–42 peptide self-induced aggregation into fibrils (by 61.7% at 10 μM) and protected cultured SH-SY5Y cells against amyloid-β-induced toxicity. These data suggest that compound 1 represents a promising candidate for hit-to-lead follow-up in the drug-discovery process against Alzheimer’s disease.

    Copyright © 2014 American Chemical Society

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. Add or change your institution or let them know you’d like them to include access.

    Supporting Information

    Click to copy section linkSection link copied!

    Results of the in silico screen for the 40 top-ranked structures from the ZINC database, FILTER configuration file, hit purity data, dose–response curves with IC50 determinations, reactivation after (±)-1-huBChE preincubation, chromatograms of chiral HPLC resolution of (±)-1, cytotoxicity profile of the virtual screening hits, X-ray crystallography data collection statistics, the comparison of the docked and crystal structure pose of compound 1, and AChE molecular docking studies. This material is available free of charge via the Internet at http://pubs.acs.org

    Accession Codes

    Atomic coordinates and structure factors for the crystal structure of huBChE in complex with compound 1 can be accessed using PDB code 4TPK.

    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

    Cited By

    Click to copy section linkSection link copied!

    This article is cited by 232 publications.

    1. Hossein Mousavi, Mehdi Rimaz, Behzad Zeynizadeh. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chemical Neuroscience 2024, 15 (9) , 1828-1881. https://doi.org/10.1021/acschemneuro.4c00055
    2. Beata Michalska, Marek Dzięgielewski, Justyna Godyń, Tobias Werner, Marek Bajda, Tadeusz Karcz, Katarzyna Szczepańska, Holger Stark, Anna Więckowska, Krzysztof Walczyński, Marek Staszewski. 4-Oxypiperidine Ethers as Multiple Targeting Ligands at Histamine H3 Receptors and Cholinesterases. ACS Chemical Neuroscience 2024, 15 (6) , 1206-1218. https://doi.org/10.1021/acschemneuro.3c00800
    3. Dang Xuan Du, Nguyen Huu Duy Khang, Nguyen Huu Tri, Pham Cam Nam, Nguyen Minh Thong. Exploring the Multitarget Activity of Wedelolactone against Alzheimer’s Disease: Insights from In Silico Study. ACS Omega 2023, 8 (17) , 15031-15040. https://doi.org/10.1021/acsomega.2c08014
    4. Muhammad Umair, Aziz ur Rehman, Muhammad Athar Abbasi, Sabahat Zahra Siddiqui, Javed Iqbal, Hira Khalid, Shahid Rasool, Shafi Ullah Khan, Fatiqa Zafar. Modular and Computational Access to Innocuous Multistep Metal-Free Synthesis of 1,3,4-Oxadiazoles as Enzyme Inhibitors. ACS Omega 2023, 8 (13) , 11952-11965. https://doi.org/10.1021/acsomega.2c07612
    5. Chenxi Du, Lei Wang, Qianwen Guan, Hongyu Yang, Tingkai Chen, Yijun Liu, Qihang Li, Weiping Lyu, Xin Lu, Ying Chen, Yang Liu, Hui Liu, Feng Feng, Wenyuan Liu, Zongliang Liu, Wei Li, Yao Chen, Haopeng Sun. N-Benzyl Benzamide Derivatives as Selective Sub-Nanomolar Butyrylcholinesterase Inhibitors for Possible Treatment in Advanced Alzheimer’s Disease. Journal of Medicinal Chemistry 2022, 65 (16) , 11365-11387. https://doi.org/10.1021/acs.jmedchem.2c00944
    6. Pedro Brandão, Óscar López, Luisa Leitzbach, Holger Stark, José G. Fernández-Bolaños, Anthony J. Burke, Marta Pineiro. Ugi Reaction Synthesis of Oxindole–Lactam Hybrids as Selective Butyrylcholinesterase Inhibitors. ACS Medicinal Chemistry Letters 2021, 12 (11) , 1718-1725. https://doi.org/10.1021/acsmedchemlett.1c00344
    7. Martina Bortolami, Fabiana Pandolfi, Valeria Tudino, Antonella Messore, Valentina Noemi Madia, Daniela De Vita, Roberto Di Santo, Roberta Costi, Isabella Romeo, Stefano Alcaro, Marisa Colone, Annarita Stringaro, Alba Espargaró, Raimon Sabatè, Luigi Scipione. New Pyrimidine and Pyridine Derivatives as Multitarget Cholinesterase Inhibitors: Design, Synthesis, and In Vitro and In Cellulo Evaluation. ACS Chemical Neuroscience 2021, 12 (21) , 4090-4112. https://doi.org/10.1021/acschemneuro.1c00485
    8. Qi Li, Ying Chen, Shuaishuai Xing, Qinghong Liao, Baichen Xiong, Yuanyuan Wang, Weixuan Lu, Siyu He, Feng Feng, Wenyuan Liu, Yao Chen, Haopeng Sun. Highly Potent and Selective Butyrylcholinesterase Inhibitors for Cognitive Improvement and Neuroprotection. Journal of Medicinal Chemistry 2021, 64 (10) , 6856-6876. https://doi.org/10.1021/acs.jmedchem.1c00167
    9. Elisabet Viayna, Nicolas Coquelle, Monika Cieslikiewicz-Bouet, Pedro Cisternas, Carolina A. Oliva, Elena Sánchez-López, Miren Ettcheto, Manuela Bartolini, Angela De Simone, Mattia Ricchini, Marisa Rendina, Mégane Pons, Omidreza Firuzi, Belén Pérez, Luciano Saso, Vincenza Andrisano, Florian Nachon, Xavier Brazzolotto, Maria Luisa García, Antoni Camins, Israel Silman, Ludovic Jean, Nibaldo C. Inestrosa, Jacques-Philippe Colletier, Pierre-Yves Renard, Diego Muñoz-Torrero. Discovery of a Potent Dual Inhibitor of Acetylcholinesterase and Butyrylcholinesterase with Antioxidant Activity that Alleviates Alzheimer-like Pathology in Old APP/PS1 Mice. Journal of Medicinal Chemistry 2021, 64 (1) , 812-839. https://doi.org/10.1021/acs.jmedchem.0c01775
    10. Jared A. Miles, Benjamin P. Ross. Recent Advances in Virtual Screening for Cholinesterase Inhibitors. ACS Chemical Neuroscience 2021, 12 (1) , 30-41. https://doi.org/10.1021/acschemneuro.0c00627
    11. Satish N. Dighe, Mangapathiraju Tippana, Suzannah van Akker, Trudi A. Collet. Structure-Based Scaffold Repurposing toward the Discovery of Novel Cholinesterase Inhibitors. ACS Omega 2020, 5 (48) , 30971-30979. https://doi.org/10.1021/acsomega.0c03848
    12. Qi Li, Shuaishuai Xing, Ying Chen, Qinghong Liao, Baichen Xiong, Siyu He, Weixuan Lu, Yang Liu, Hongyu Yang, Qihang Li, Feng Feng, Wenyuan Liu, Yao Chen, Haopeng Sun. Discovery and Biological Evaluation of a Novel Highly Potent Selective Butyrylcholinsterase Inhibitor. Journal of Medicinal Chemistry 2020, 63 (17) , 10030-10044. https://doi.org/10.1021/acs.jmedchem.0c01129
    13. Yong-Qiang Li, Li-Sha Bai, Cai-Xia Zhao, Jing-Jing Xu, Zhong-Juan Sun, Yan-Ling Dong, De-Xian Li, Xi-Li Liu, Zhi-Qing Ma. Functional Characterization of Two Carboxylesterase Genes Involved in Pyrethroid Detoxification in Helicoverpa armigera. Journal of Agricultural and Food Chemistry 2020, 68 (11) , 3390-3402. https://doi.org/10.1021/acs.jafc.9b06690
    14. Masha G. Savelieff, Geewoo Nam, Juhye Kang, Hyuck Jin Lee, Misun Lee, Mi Hee Lim. Development of Multifunctional Molecules as Potential Therapeutic Candidates for Alzheimer’s Disease, Parkinson’s Disease, and Amyotrophic Lateral Sclerosis in the Last Decade. Chemical Reviews 2019, 119 (2) , 1221-1322. https://doi.org/10.1021/acs.chemrev.8b00138
    15. Dawid Panek, Anna Więckowska, Jakub Jończyk, Justyna Godyń, Marek Bajda, Tomasz Wichur, Anna Pasieka, Damijan Knez, Anja Pišlar, Jan Korabecny, Ondrej Soukup, Vendula Sepsova, Raimon Sabaté, Janko Kos, Stanislav Gobec, Barbara Malawska. Design, Synthesis, and Biological Evaluation of 1-Benzylamino-2-hydroxyalkyl Derivatives as New Potential Disease-Modifying Multifunctional Anti-Alzheimer’s Agents. ACS Chemical Neuroscience 2018, 9 (5) , 1074-1094. https://doi.org/10.1021/acschemneuro.7b00461
    16. Urban Košak, Boris Brus, Damijan Knez, Simon Žakelj, Jurij Trontelj, Anja Pišlar, Roman Šink, Marko Jukič, Marko Živin, Adrian Podkowa, Florian Nachon, Xavier Brazzolotto, Jure Stojan, Janko Kos, Nicolas Coquelle, Kinga Sałat, Jacques-Philippe Colletier, and Stanislav Gobec . The Magic of Crystal Structure-Based Inhibitor Optimization: Development of a Butyrylcholinesterase Inhibitor with Picomolar Affinity and in Vivo Activity. Journal of Medicinal Chemistry 2018, 61 (1) , 119-139. https://doi.org/10.1021/acs.jmedchem.7b01086
    17. Srinivas Kantham, Stephen Chan, Gawain McColl, Jared A. Miles, Suresh Kumar Veliyath, Girdhar Singh Deora, Satish N. Dighe, Samira Khabbazi, Marie-Odile Parat, and Benjamin P. Ross . Effect of the Biphenyl Neolignan Honokiol on Aβ42-Induced Toxicity in Caenorhabditis elegans, Aβ42 Fibrillation, Cholinesterase Activity, DPPH Radicals, and Iron(II) Chelation. ACS Chemical Neuroscience 2017, 8 (9) , 1901-1912. https://doi.org/10.1021/acschemneuro.7b00071
    18. Giulio Vistoli, Angelica Mazzolari, Bernard Testa, and Alessandro Pedretti . Binding Space Concept: A New Approach To Enhance the Reliability of Docking Scores and Its Application to Predicting Butyrylcholinesterase Hydrolytic Activity. Journal of Chemical Information and Modeling 2017, 57 (7) , 1691-1702. https://doi.org/10.1021/acs.jcim.7b00121
    19. Satish N. Dighe, Girdhar Singh Deora, Eugenio De la Mora, Florian Nachon, Stephen Chan, Marie-Odile Parat, Xavier Brazzolotto, and Benjamin P. Ross . Discovery and Structure–Activity Relationships of a Highly Selective Butyrylcholinesterase Inhibitor by Structure-Based Virtual Screening. Journal of Medicinal Chemistry 2016, 59 (16) , 7683-7689. https://doi.org/10.1021/acs.jmedchem.6b00356
    20. Serena Montanari, Laura Scalvini, Manuela Bartolini, Federica Belluti, Silvia Gobbi, Vincenza Andrisano, Alessia Ligresti, Vincenzo Di Marzo, Silvia Rivara, Marco Mor, Alessandra Bisi, and Angela Rampa . Fatty Acid Amide Hydrolase (FAAH), Acetylcholinesterase (AChE), and Butyrylcholinesterase (BuChE): Networked Targets for the Development of Carbamates as Potential Anti-Alzheimer’s Disease Agents. Journal of Medicinal Chemistry 2016, 59 (13) , 6387-6406. https://doi.org/10.1021/acs.jmedchem.6b00609
    21. Javier E. Ortiz, Natalia B. Pigni, Sebastián A. Andujar, German Roitman, Fernando D. Suvire, Ricardo D. Enriz, Alejandro Tapia, Jaume Bastida, and Gabriela E. Feresin . Alkaloids from Hippeastrum argentinum and Their Cholinesterase-Inhibitory Activities: An in Vitro and in Silico Study. Journal of Natural Products 2016, 79 (5) , 1241-1248. https://doi.org/10.1021/acs.jnatprod.5b00785
    22. Edgar Sawatzky, Sarah Wehle, Beata Kling, Jan Wendrich, Gerhard Bringmann, Christoph A. Sotriffer, Jörg Heilmann, and Michael Decker . Discovery of Highly Selective and Nanomolar Carbamate-Based Butyrylcholinesterase Inhibitors by Rational Investigation into Their Inhibition Mode. Journal of Medicinal Chemistry 2016, 59 (5) , 2067-2082. https://doi.org/10.1021/acs.jmedchem.5b01674
    23. Peng Zhan, Yukihiro Itoh, Takayoshi Suzuki, and Xinyong Liu . Strategies for the Discovery of Target-Specific or Isoform-Selective Modulators. Journal of Medicinal Chemistry 2015, 58 (19) , 7611-7633. https://doi.org/10.1021/acs.jmedchem.5b00229
    24. Samo Lešnik, Tanja Štular, Boris Brus, Damijan Knez, Stanislav Gobec, Dušanka Janežič, and Janez Konc . LiSiCA: A Software for Ligand-Based Virtual Screening and Its Application for the Discovery of Butyrylcholinesterase Inhibitors. Journal of Chemical Information and Modeling 2015, 55 (8) , 1521-1528. https://doi.org/10.1021/acs.jcim.5b00136
    25. Riham A. El-Shiekh, Dalia E. Ali, Asmaa A. Mandour, Meselhy R. Meselhy. Essential oils of Plumeria alba L. and Plumeria rubra L. growing in Egypt: GC/MS analysis, molecular dynamics and in-vitro anti-cholinesterase activity. Industrial Crops and Products 2024, 221 , 119316. https://doi.org/10.1016/j.indcrop.2024.119316
    26. Chang Xu, Xiao Yu, Guiyang Wang, Shiqing You, Linlin Zhu, Ying Liu, Nuan Zhang, Zhengdong Wang, Bin Liu, Wei Zhang. Affinity ultrafiltration based metabolomic profiling directed discovery novel butyrylcholinesterase inhibitors from Uncaria sessilifructus. International Journal of Biological Macromolecules 2024, 277 , 134179. https://doi.org/10.1016/j.ijbiomac.2024.134179
    27. Marie Nevyhoštěná, Alena Komersová, Vladimír Pejchal, Šárka Štěpánková, Petr Česla, Kevin Matzick, Jana Macháčková, Roman Svoboda. New biologically active sulfonamides as potential drugs for Alzheimer's disease. Archiv der Pharmazie 2024, 357 (10) https://doi.org/10.1002/ardp.202400191
    28. Xiao-Xing Zhu, Feng Gao, Lin-Xi Wan. Design, synthesis and anti-Alzheimer's disease activity evaluation of C-3 arylated huperzine A derivatives. Fitoterapia 2024, 177 , 106141. https://doi.org/10.1016/j.fitote.2024.106141
    29. Hatim Soufi, Mohamed Moussaoui, Soukayna Baammi, Mouna Baassi, Mohammed Salah, Rachid Daoud, Achraf El Allali, M. E. Belghiti, Mohammed Moutaabbid, Said Belaaouad. Multi-combined QSAR, molecular docking, molecular dynamics simulation, and ADMET of Flavonoid derivatives as potent cholinesterase inhibitors. Journal of Biomolecular Structure and Dynamics 2024, 42 (12) , 6027-6041. https://doi.org/10.1080/07391102.2023.2238314
    30. Jing. Pei Zou, Xiao. Qing Zhang, Qian. Wen Guo, Xu. Hui Xu, Wei. Wei Liu, Feng Zhang, Da. Hua Shi. Synthesis, biological activity, X-ray crystallographic, DFT calculations and molecular dynamics simulation studies of 2-phenylthiazole-1,3,5-triazine derivatives as potential cholinesterase inhibitors. Journal of Molecular Structure 2024, 1309 , 138203. https://doi.org/10.1016/j.molstruc.2024.138203
    31. Ebru Koçak Aslan, Aysima Sezer, Tuba Tüylü Küçükkılınç, Erhan Palaska. Novel 1,2,4‐triazole derivatives containing the naphthalene moiety as selective butyrylcholinesterase inhibitors: Design, synthesis, and biological evaluation. Archiv der Pharmazie 2024, 51 https://doi.org/10.1002/ardp.202400406
    32. Zhen-Qing Yu, Yongsheng Ma, Shuai Xu, Lei Yang, Yongqing Zhou, Xiaofeng Yang, Xiuqi Kong, Yanfeng Lv, Jing Zhang, Mei Yan. Bioinspired design of highly specific fluorescent probe for butyrylcholinesterase imaging in living cells and Alzheimer's disease model. Sensors and Actuators B: Chemical 2024, 410 , 135662. https://doi.org/10.1016/j.snb.2024.135662
    33. Musa Özil, Halis T. Balaydın, Berna Dogan, Murat Şentürk, Serdar Durdagi. Efficient, rapid, and high‐yield synthesis of aryl Schiff base derivatives and their in vitro and in silico inhibition studies of hCA I, hCA II, AChE, and BuChE. Archiv der Pharmazie 2024, 357 (7) https://doi.org/10.1002/ardp.202300266
    34. Ziwei Guo, Junqian Peng, Zhiqiang Zhou, Fang Wang, Mingfang He, Sheng Lu, Xiaoqiang Chen. Benzorhodol derived far-red/near-infrared fluorescent probes for selective and sensitive detection of butyrylcholinesterase activity in living cells and the non-alcoholic fatty liver of zebrafish. Analytical Methods 2024, 16 (25) , 4054-4059. https://doi.org/10.1039/D4AY00662C
    35. Petar Batinić, Aleksandra Jovanović, Dejan Stojković, Natalija Čutović, Ilija Cvijetić, Uroš Gašić, Tamara Carević, Gökhan Zengin, Aleksandar Marinković, Tatjana Marković. A novel source of biologically active compounds – The leaves of Serbian herbaceous peonies. Saudi Pharmaceutical Journal 2024, 32 (6) , 102090. https://doi.org/10.1016/j.jsps.2024.102090
    36. Lisa Sequeira, Sofia Benfeito, Carlos Fernandes, Inês Lima, Joana Peixoto, Catarina Alves, Cláudia Sofia Machado, Alexandra Gaspar, Fernanda Borges, Daniel Chavarria. Drug Development for Alzheimer’s and Parkinson’s Disease: Where Do We Go Now?. Pharmaceutics 2024, 16 (6) , 708. https://doi.org/10.3390/pharmaceutics16060708
    37. Luyi Ooi, Kim San Tang, Joash Ban Lee Tan, Keng Yoon Yeong. The Discovery of Benzylidene‐Indenone as Selective Butyrylcholinesterase Inhibitor with Choline Acetyltransferase Gene and Neurite Promoting Abilities. ChemistrySelect 2024, 9 (19) https://doi.org/10.1002/slct.202400695
    38. Ayesha Tahir, Bushra Mobeen, Fahad Hussain, Abdul Sadiq, Umer Rashid. Pregnenolone derivatives for the treatment of Alzheimer's disease: synthesis, and in vitro inhibition of amyloid β 1–42 peptide aggregation, acetylcholinesterase and carbonic anhydrase-II. RSC Advances 2024, 14 (21) , 14742-14757. https://doi.org/10.1039/D4RA01536C
    39. Petar Batinić, Aleksandra Jovanović, Dejan Stojković, Gökhan Zengin, Ilija Cvijetić, Uroš Gašić, Natalija Čutović, Mirjana B. Pešić, Danijel D. Milinčić, Tamara Carević, Aleksandar Marinković, Branko Bugarski, Tatjana Marković. Phytochemical Analysis, Biological Activities, and Molecular Docking Studies of Root Extracts from Paeonia Species in Serbia. Pharmaceuticals 2024, 17 (4) , 518. https://doi.org/10.3390/ph17040518
    40. Ying Chen, Weiting Zhang, Qi Li, Huanfang Xie, Shuaishuai Xing, Xin Lu, Weiping Lyu, Baichen Xiong, Yuanyuan Wang, Wei Qu, Wenyuan Liu, Heng Chi, Xiaolong Zhang, Feng Feng, Haopeng Sun. Discovery of 4-benzylpiperazinequinoline BChE inhibitor that suppresses neuroinflammation for the treatment of Alzheimer's Disease. European Journal of Medicinal Chemistry 2024, 10 , 116463. https://doi.org/10.1016/j.ejmech.2024.116463
    41. Hamideh Dehghani, Marzieh Rashedinia, Gholamhossein Mohebbi, Amir Vazirizadeh. Studies on Secondary Metabolites and In vitro and In silico Anticholinesterases Activities of the Sea Urchin Echinometra mathaei Crude Venoms from the Persian Gulf-Bushehr. The Natural Products Journal 2024, 14 (2) https://doi.org/10.2174/2210315514666230622144244
    42. Ali Naderi, Akın Akıncıoğlu, Ahmet Çağan, Hilal Çelikkaleli, Hülya Akıncıoğlu, Süleyman Göksu. Design, synthesis and anticholinergic properties of novel α-benzyl dopamine, tyramine, and phenethylamine derivatives. Bioorganic Chemistry 2024, 144 , 107146. https://doi.org/10.1016/j.bioorg.2024.107146
    43. Shuaishuai Xing, Xu Tang, Leyan wang, Jun Wang, Bingbing Lv, Xiaolong Wang, Can Guo, Ye Zhao, Feng Feng, Wenyuan Liu, Yao Chen, Haopeng Sun. Optimizing drug-like properties of selective butyrylcholinesterase inhibitors for cognitive improvement: Enhancing aqueous solubility by disrupting molecular plane. European Journal of Medicinal Chemistry 2024, 268 , 116289. https://doi.org/10.1016/j.ejmech.2024.116289
    44. Leo Bon, Angelika Banaś, Inês Dias, Inês Melo-Marques, Sandra M. Cardoso, Sílvia Chaves, M. Amélia Santos. New Multitarget Rivastigmine–Indole Hybrids as Potential Drug Candidates for Alzheimer’s Disease. Pharmaceutics 2024, 16 (2) , 281. https://doi.org/10.3390/pharmaceutics16020281
    45. Xiao‐Qing Zhang, Jing Wang, Jing‐Pei Zou, Yang Cao, Xu‐Hui Xu, Bo Ding, Wei‐Wei Liu, Shao‐Jie Ma, Da‐Hua Shi. Design, Synthesis and Anticholinesterase Activity of Coumarin‐1,3,5‐triazine Derivatives. ChemistrySelect 2024, 9 (3) https://doi.org/10.1002/slct.202303428
    46. Ani S. Dadayan, Anna F. Mkrtchyan, Atravazd S. Poghosyan, Slavik A. Dadayan, Lala A. Stepanyan, Monika H. Israyelyan, Anna S. Tovmasyan, Avetis H. Tsaturyan, Nona A. Hovhannisyan, Vigen O. Topuzyan, Tatevik H. Sargsyan, Ashot S. Saghyan. Unnatural Phosphorus‐Containing α‐Amino Acids and Their N‐FMOC Derivatives: Synthesis and In Vitro Investigation of Anticholinesterase Activity. ChemistrySelect 2024, 9 (3) https://doi.org/10.1002/slct.202303249
    47. Kashif Maqbool Khan, Muhammad Faisal Nadeem, Abdul Mannan, Tahir Ali Chohan, Muhammad Islam, Siddique Akber Ansari, Hamad M. Alkahtani, Irfan Aamer Ansari, Umair Khurshid, Syafiq Asnawi Zainal Abidin, Syed Adnan Ali Shah, Hammad Saleem. Biochemical, Toxicological, and in Silico Aspects of Trillium govanianum Wall. ex D.Don (Trilliaceae): A Rich Source of Natural Bioactive Compounds. Chemistry & Biodiversity 2024, 21 (1) https://doi.org/10.1002/cbdv.202301375
    48. Devika Sonawane, Varsha Pokharkar. A Comparative Study of Binding Interactions of Natural Flavonoids and Conventional Drug Donepezil for Multiple Alzheimer's Disease Targets Using In silico Approach. Letters in Drug Design & Discovery 2023, 20 (12) , 1947-1958. https://doi.org/10.2174/1570180819666220509092139
    49. Roghayeh Esfandiari, Parsa Moghimi-Rad, Mohammed Hussen Bule, Effat Souri, Hamid Nadri, Mohammad Mahdavi, Roshanak Ghobadian, Mohsen Amini. New Imidazo[1,2-a]pyridin-2-yl Derivatives as AChE, BChE, and LOX Inhibitors; Design, Synthesis, and Biological Evaluation. Letters in Drug Design & Discovery 2023, 20 (11) , 1784-1798. https://doi.org/10.2174/1570180819666220608111906
    50. Raissa Alves da Conceição, Natalia von Ranke, Luciana Azevedo, Daiana Franco, Nathalia Fonseca Nadur, Arthur Eugen Kummerle, Maria Letícia de C. Barbosa, Alessandra M. T. Souza. Structure‐based design of new N ‐benzyl‐piperidine derivatives as multitarget‐directed AChE/BuChE inhibitors for Alzheimer's disease. Journal of Cellular Biochemistry 2023, 124 (11) , 1734-1748. https://doi.org/10.1002/jcb.30483
    51. Fatemeh Pishgouii, Safa Lotfi, Ebrahim Sedaghati. Anti-AChE and Anti-BuChE Screening of the Fermentation Broth Extracts from Twelve Aspergillus Isolates and GC-MS and Molecular Docking Studies of the Most Active Extracts. Applied Biochemistry and Biotechnology 2023, 195 (8) , 5199-5216. https://doi.org/10.1007/s12010-023-04548-0
    52. Shahnaz Fathi Hafshejani, Safa Lotfi, Elham Rezvannejad, Mojtaba Mortazavi, Ali Riahi‐Madvar. Correlation between total phenolic and flavonoid contents and biological activities of 12 ethanolic extracts of Iranian propolis. Food Science & Nutrition 2023, 11 (7) , 4308-4325. https://doi.org/10.1002/fsn3.3356
    53. Filip Pidany, Jana Kroustkova, Abdullah Al Mamun, Daniela Suchankova, Xavier Brazzolotto, Florian Nachon, Fabien Chantegreil, Rafael Dolezal, Lenka Pulkrabkova, Lubica Muckova, Martina Hrabinova, Vladimir Finger, Martin Kufa, Ondrej Soukup, Daniel Jun, Jaroslav Jenco, Jiri Kunes, Lucie Novakova, Jan Korabecny, Lucie Cahlikova. Highly selective butyrylcholinesterase inhibitors related to Amaryllidaceae alkaloids - Design, synthesis, and biological evaluation. European Journal of Medicinal Chemistry 2023, 252 , 115301. https://doi.org/10.1016/j.ejmech.2023.115301
    54. Sunil Kumar, Amritha Manoharan, Jayalakshmi J, Mohamed A. Abdelgawad, Wael A. Mahdi, Sultan Alshehri, Mohammed M. Ghoneim, Leena K. Pappachen, Subin Mary Zachariah, T. P. Aneesh, Bijo Mathew. Exploiting butyrylcholinesterase inhibitors through a combined 3-D pharmacophore modeling, QSAR, molecular docking, and molecular dynamics investigation. RSC Advances 2023, 13 (14) , 9513-9529. https://doi.org/10.1039/D3RA00526G
    55. Rubina Munir, Sumera Zaib, Muhammad Zia-ur-Rehman, Nadia Hussain, Faryal Chaudhry, Muhammad Tayyab Younas, Fatima Tuz Zahra, Zainab Tajammul, Noman Javid, Ayed A. Dera, Hanan A. Ogaly, Imtiaz Khan. Ultrasound-Assisted Synthesis of Piperidinyl-Quinoline Acylhydrazones as New Anti-Alzheimer’s Agents: Assessment of Cholinesterase Inhibitory Profile, Molecular Docking Analysis, and Drug-like Properties. Molecules 2023, 28 (5) , 2131. https://doi.org/10.3390/molecules28052131
    56. Łukasz Pecio, Mostafa Alilou, Solomiia Kozachok, Ilkay Erdogan Orhan, Gökçen Eren, Fatma Sezer Şenol Deniz, Hermann Stuppner, Wiesław Oleszek. Absolute configuration of spiro-flavostilbenoids from Yucca schidigera Roezl ex Ortgies: First indication of (2R)-naringenin as the key building block. Phytochemistry 2023, 207 , 113584. https://doi.org/10.1016/j.phytochem.2022.113584
    57. Sarfaraz Shaikh, Pratik Dhavan, Pinky Singh, Jasmin Uparkar, S. P. Vaidya, B. L. Jadhav, M. M. V. Ramana. Design, synthesis and biological evaluation of novel antipyrine based α-aminophosphonates as anti-Alzheimer and anti-inflammatory agent. Journal of Biomolecular Structure and Dynamics 2023, 41 (2) , 386-401. https://doi.org/10.1080/07391102.2021.2006088
    58. Zhipei Sang, Ling Huang. Alzheimer's disease therapeutics. 2023, 405-473. https://doi.org/10.1016/B978-0-443-18611-0.00031-0
    59. Yiqun Chang, Bryson A. Hawkins, Jonathan J. Du, Paul W. Groundwater, David E. Hibbs, Felcia Lai. A Guide to In Silico Drug Design. Pharmaceutics 2023, 15 (1) , 49. https://doi.org/10.3390/pharmaceutics15010049
    60. Shuang Miao, Qianqian He, Chen Li, Yan Wu, Mengshan Liu, Yongshou Chen, Shizhou Qi, Kaikai Gong. Aaptamine – a dual acetyl – and butyrylcholinesterase inhibitor as potential anti-Alzheimer’s disease agent. Pharmaceutical Biology 2022, 60 (1) , 1502-1510. https://doi.org/10.1080/13880209.2022.2102657
    61. Xiang Cui, Shanshan Deng, Guoyin Li, Yunxia Zhang, Lining Wang, Changjing Wu, Yanru Deng. Butenolide derivatives from Aspergillus terreus selectively inhibit butyrylcholinesterase. Frontiers in Chemistry 2022, 10 https://doi.org/10.3389/fchem.2022.1063284
    62. Deepak Mishra, Atiya Fatima, Prashant Kumar, Nupur S Munjal, Brajendra K Singh, Ram Singh. Synthesis of Benzothiazole Linked Triazole Conjugates and Their Evaluation Against Cholinesterase Enzymes. ChemistrySelect 2022, 7 (46) https://doi.org/10.1002/slct.202203060
    63. Shouyuan Sun, Tao Shi, Yan Peng, Honghua Zhang, Linsheng Zhuo, Xue Peng, Qien Li, Manxia Wang, Shuzhi Wang, Zhen Wang. Discovery of pyrrole derivatives as acetylcholinesterase-sparing butyrylcholinesterase inhibitor. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.1043397
    64. Mohammad Hadi Meshkatalsadat, Ahmad Mahmoudi, Safa Lotfi, Behjat Pouramiri, Alireza Foroumadi. Green and four-component cyclocondensation synthesis and in silico docking of new polyfunctionalized pyrrole derivatives as the potential anticholinesterase agents. Molecular Diversity 2022, 26 (6) , 3021-3035. https://doi.org/10.1007/s11030-021-10362-9
    65. Yi Liu, Yanbei Tu, Yunyao Kang, Chao Zhu, Chuanhai Wu, Gang Chen, Zerong Liu, Yanfang Li. Biological evaluation, molecular modeling and dynamics simulation of phenanthrenes isolated from Bletilla striata as butyrylcholinesterase inhibitors. Scientific Reports 2022, 12 (1) https://doi.org/10.1038/s41598-022-17912-7
    66. Yuying Wang, Lin Long, Quanwei Yu, Honghua Zhang, Xuelin Li, Linsheng Zhuo, Shuzhi Wang, Zhen Wang. Discovery of carbamate-based Salicylic acid derivatives as novel Cholinesterase inhibitor. Journal of Molecular Structure 2022, 397 , 134804. https://doi.org/10.1016/j.molstruc.2022.134804
    67. Ilkay Erdogan Orhan, F. Sezer Senol Deniz, Ramin Ekhteiari Salmas, Sule Irmak, Ozden Ozgun Acar, Gurbet Celik Turgut, Alaattin Sen, Ana‐Maria Zbancioc, Simon Vlad Luca, Adrianna Skiba, Krystyna Skalicka‐Woźniak, Gabriela Tataringa. Evaluation of Anti‐Alzheimer Activity of Synthetic Coumarins by Combination of in Vitro and in Silico Approaches. Chemistry & Biodiversity 2022, 19 (12) https://doi.org/10.1002/cbdv.202200315
    68. Wen-da Zhang, Jing-min Zhang, Chong-zhen Qin, Xin-ru Wang, Yu-bing Zhou. A far-red/near-infrared fluorescence probe with large Stokes shift for monitoring butyrylcholinesterase (BChE) in living cells and in vivo. Analytica Chimica Acta 2022, 1235 , 340540. https://doi.org/10.1016/j.aca.2022.340540
    69. Marcela C. R. Silva, Adriana F. L. Vilela, Carmen L. Cardoso, Ronaldo A. Pilli. Synthesis and Anticholinesterase Evaluation of Cassine, Spectaline and Analogues. Scientia Pharmaceutica 2022, 90 (4) , 63. https://doi.org/10.3390/scipharm90040063
    70. Junbo Wu, Honghua Zhang, Yuying Wang, Gaofeng Yin, Qien Li, Linsheng Zhuo, Hongjin Chen, Zhen Wang. From tryptamine to the discovery of efficient multi-target directed ligands against cholinesterase-associated neurodegenerative disorders. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.1036030
    71. Fatima Y. Adeowo, Ahmed A. Elrashedy, Murtala A. Ejalonibu, Isiaka A. Lawal, Monsurat M. Lawal, Hezekiel M. Kumalo. Pharmacophore mapping of the crucial mediators of acetylcholinesterase and butyrylcholinesterase dual inhibition in Alzheimer's disease. Molecular Diversity 2022, 26 (5) , 2761-2774. https://doi.org/10.1007/s11030-022-10377-w
    72. Yuexia Yang, Liang Zhang, Jiemin Wang, Yuping Cao, Wenwu Qin, Yun Liu. Real-time fluorescent determination and biological imaging in living models via a butyrylcholinesterase-activated fluorescent probe. Dyes and Pigments 2022, 206 , 110596. https://doi.org/10.1016/j.dyepig.2022.110596
    73. Sabrina Lekmine, Samira Bendjedid, Ouided Benslama, Antonio Ignacio Martín-García, Samira Boussekine, Kenza Kadi, Salah Akkal, Gema Nieto, Rokayya Sami, Amina A. M. Al-Mushhin, Morooj M. Baakdah, Abeer M. Aljaadi, Saif A. Alharthy. Ultrasound-Assisted Extraction, LC–MS/MS Analysis, Anticholinesterase, and Antioxidant Activities of Valuable Natural Metabolites from Astragalus armatus Willd.: In Silico Molecular Docking and In Vitro Enzymatic Studies. Antioxidants 2022, 11 (10) , 2000. https://doi.org/10.3390/antiox11102000
    74. Ante Miličević, Goran Šinko. Evaluation of the Key Structural Features of Various Butyrylcholinesterase Inhibitors Using Simple Molecular Descriptors. Molecules 2022, 27 (20) , 6894. https://doi.org/10.3390/molecules27206894
    75. Ali akbar Mariki, Akbar Anaeigoudari, Mahboobeh Zahedifar, Behjat Pouramiri, Adileh Ayati, Safa Lotfi. Design, Green Synthesis, and Biological Evaluation of New Substituted Tetrahydropyrimidine Derivatives as Acetylcholinesterase Inhibitors. Polycyclic Aromatic Compounds 2022, 42 (8) , 5231-5241. https://doi.org/10.1080/10406638.2021.1933102
    76. Jaromir Myslivecek. Multitargeting nature of muscarinic orthosteric agonists and antagonists. Frontiers in Physiology 2022, 13 https://doi.org/10.3389/fphys.2022.974160
    77. Mehmet Abdullah Alagöz, Seong-Min Kim, Jong Min Oh, Gülnur Arslan, Zeynep Özdemir, Suat Sari, Azime Berna Özçelik, Tijen Önkol, Daniela Trisciuzzi, Orazio Nicolotti, Hoon Kim, Bijo Mathew. Inhibition of Cholinesterases by Benzothiazolone Derivatives. Processes 2022, 10 (9) , 1872. https://doi.org/10.3390/pr10091872
    78. Sarfaraz Shaikh, Pratik Dhavan, Pinky Singh, Jasmin Uparkar, S. P. Vaidya, B. L. Jadhav, M. V. Ramana. Synthesis of carbazole based α-aminophosphonate derivatives: design, molecular docking and in vitro cholinesterase activity. Journal of Biomolecular Structure and Dynamics 2022, 40 (11) , 4801-4814. https://doi.org/10.1080/07391102.2020.1861981
    79. David Vicente-Zurdo, Noelia Rosales-Conrado, M. Eugenia León-González, Leonardo Brunetti, Luca Piemontese, A. Raquel Pereira-Santos, Sandra M. Cardoso, Yolanda Madrid, Sílvia Chaves, M. Amélia Santos. Novel Rivastigmine Derivatives as Promising Multi-Target Compounds for Potential Treatment of Alzheimer’s Disease. Biomedicines 2022, 10 (7) , 1510. https://doi.org/10.3390/biomedicines10071510
    80. Zhenqing Yu, Xiang Li, Xiaofeng Lu, Yong Guo. Rational construction of a novel probe for the rapid detection of butyrylcholinesterase stress changes in apoptotic cells. New Journal of Chemistry 2022, 46 (25) , 12034-12040. https://doi.org/10.1039/D2NJ01678H
    81. Martina Bortolami, Fabiana Pandolfi, Valeria Tudino, Antonella Messore, Valentina Noemi Madia, Daniela De Vita, Roberto Di Santo, Roberta Costi, Isabella Romeo, Stefano Alcaro, Marisa Colone, Annarita Stringaro, Alba Espargaró, Raimon Sabatè, Luigi Scipione. Design, Synthesis, and In Vitro, In Silico and In Cellulo Evaluation of New Pyrimidine and Pyridine Amide and Carbamate Derivatives as Multi-Functional Cholinesterase Inhibitors. Pharmaceuticals 2022, 15 (6) , 673. https://doi.org/10.3390/ph15060673
    82. Meiyang Xi, Chengjie Feng, Kui Du, Weiping Lv, Chenxi Du, Runpu Shen, Haopeng Sun. Design, synthesis, biological evaluation and molecular modeling of N-isobutyl-N-((2-(p-tolyloxymethyl)thiazol-4yl)methyl)benzo[d][1,3] dioxole-5-carboxamides as selective butyrylcholinesterase inhibitors. Bioorganic & Medicinal Chemistry Letters 2022, 61 , 128602. https://doi.org/10.1016/j.bmcl.2022.128602
    83. Mihajlo Gajic, Damijan Knez, Izidor Sosič, Janez Mravljak, Anže Meden, Urban Košak, Luisa Leitzbach, Sven George, Bettina Hofmann, Aleksandra Zivkovic, Dieter Steinhilber, Holger Stark, Stanislav Gobec, Andrija Smelcerovic, Marko Anderluh. Repurposing of 8‐Hydroxyquinoline‐Based Butyrylcholinesterase and Cathepsin B Ligands as Potent Nonpeptidic Deoxyribonuclease I Inhibitors. ChemMedChem 2022, 17 (5) https://doi.org/10.1002/cmdc.202100694
    84. José David Sánchez-Martínez, Gerardo Alvarez-Rivera, Rocio Gallego, Mariane Bittencourt Fagundes, Alberto Valdés, Jose A. Mendiola, Elena Ibañez, Alejandro Cifuentes. Neuroprotective potential of terpenoid-rich extracts from orange juice by-products obtained by pressurized liquid extraction. Food Chemistry: X 2022, 13 , 100242. https://doi.org/10.1016/j.fochx.2022.100242
    85. Aso Hameed Hasan, Sankaranarayanan Murugesan, Syazwani Itri Amran, Subhash Chander, Mohammed M. Alanazi, Taibi Ben Hadda, Sonam Shakya, Mohammad Rizki Fadhil Pratama, Basundhara Das, Subhrajit Biswas, Joazaizulfazli Jamalis. Novel thiophene Chalcones-Coumarin as acetylcholinesterase inhibitors: Design, synthesis, biological evaluation, molecular docking, ADMET prediction and molecular dynamics simulation. Bioorganic Chemistry 2022, 119 , 105572. https://doi.org/10.1016/j.bioorg.2021.105572
    86. Petr Jansa, Ivan Barvík, Daniela Hulcová, Eliška Matoušová. Synthesis and cholinesterase inhibitory activity study of Amaryllidaceae alkaloid analogues with N -methyl substitution. Organic & Biomolecular Chemistry 2022, 19 https://doi.org/10.1039/D2OB00553K
    87. Mahshid Attarroshan, Omidreza Firuzi, Aida Iraji, Shahrzad Sharifi, Marjan Tavakkoli, Mahmmod Vesal, Mahsima Khoshneviszadeh, Somayeh Pirhadi, Najmeh Edraki. Imino‐2 H ‐Chromene Based Derivatives as Potential Anti‐Alzheimer's Agents: Design, Synthesis, Biological Evaluation and in Silico Study. Chemistry & Biodiversity 2022, 19 (1) https://doi.org/10.1002/cbdv.202100599
    88. Shuaizhang Li, Andrew J. Li, Jameson Travers, Tuan Xu, Srilatha Sakamuru, Carleen Klumpp-Thomas, Ruili Huang, Menghang Xia. Identification of Compounds for Butyrylcholinesterase Inhibition. SLAS Discovery 2021, 26 (10) , 1355-1364. https://doi.org/10.1177/24725552211030897
    89. Mehtap Kilic, Ilkay Erdogan Orhan, Gokcen Eren, Emine Sukran Okudan, Alden S. Estep, James J. Bencel, Nurhayat Tabanca. Insecticidal activity of forty-seven marine algae species from the Mediterranean, Aegean, and Sea of Marmara in connection with their cholinesterase and tyrosinase inhibitory activity. South African Journal of Botany 2021, 143 , 435-442. https://doi.org/10.1016/j.sajb.2021.06.038
    90. Sarfaraz Shaikh, Pratik Dhavan, Jasmin Uparkar, Pinky Singh, S.P. Vaidya, B.L. Jadhav, M.M.V. Ramana. Synthesis, characterization, in vitro cholinesterase and hRBCs hemolysis assay and computational evaluation of novel 2,3,4,5-tetrahydrobenzothiazepine appended α-aminophosphonates. Bioorganic Chemistry 2021, 116 , 105397. https://doi.org/10.1016/j.bioorg.2021.105397
    91. Negar Maafi, Filip Pidaný, Jana Maříková, Jan Korábečný, Daniela Hulcová, Tomáš Kučera, Monika Schmidt, Latifah Al Shammari, Marcel Špulák, Maria Carmen Catapano, Marko Mecava, Lukáš Prchal, Jiří Kuneš, Jiří Janoušek, Eliška Kohelová, Jaroslav Jenčo, Lucie Nováková, Lucie Cahlíková. Derivatives of montanine-type alkaloids and their implication for the treatment of Alzheimer's disease: Synthesis, biological activity and in silico study. Bioorganic & Medicinal Chemistry Letters 2021, 51 , 128374. https://doi.org/10.1016/j.bmcl.2021.128374
    92. Fatima Y. Adeowo, Tosin P. Oyetunji, Murtala A. Ejalonibu, Umar Ndagi, Hezekiel M. Kumalo, Monsurat M. Lawal. Tailored Modeling of Rivastigmine Derivatives as Dual Acetylcholinesterase and Butyrylcholinesterase Inhibitors for Alzheimer's Disease Treatment. Chemistry & Biodiversity 2021, 18 (11) https://doi.org/10.1002/cbdv.202100361
    93. Jiarui Wu, Xiaodi Kou, Hui Ju, Hongwei Zhang, Aihong Yang, Rui Shen. Design, synthesis and biological evaluation of naringenin carbamate derivatives as potential multifunctional agents for the treatment of Alzheimer’s disease. Bioorganic & Medicinal Chemistry Letters 2021, 49 , 128316. https://doi.org/10.1016/j.bmcl.2021.128316
    94. Marwa Gardelly, Belsem Trimech, Mabrouk Horchani, Mansour Znati, Hichem Ben Jannet, Anis Romdhane. Anti-tyrosinase and Anti-butyrylcholinesterase Quinolines-Based Coumarin Derivatives: Synthesis and Insights from Molecular Docking Studies. Chemistry Africa 2021, 4 (3) , 491-501. https://doi.org/10.1007/s42250-021-00235-x
    95. Hongwei Gao, Yingying Jiang, Jiuyu Zhan, Yingni Sun. Pharmacophore-based drug design of AChE and BChE dual inhibitors as potential anti-Alzheimer’s disease agents. Bioorganic Chemistry 2021, 114 , 105149. https://doi.org/10.1016/j.bioorg.2021.105149
    96. Pratibha Magar, Oscar Parravicini, Šárka Štěpánková, Katarina Svrčková, Adriana D. Garro, Izabela Jendrzejewska, Karel Pauk, Jan Hošek, Josef Jampílek, Ricardo D. Enriz, Aleš Imramovský. Novel Sulfonamide-Based Carbamates as Selective Inhibitors of BChE. International Journal of Molecular Sciences 2021, 22 (17) , 9447. https://doi.org/10.3390/ijms22179447
    97. Ilkay Erdogan Orhan, Fatma Tosun, Fatma Sezer Senol Deniz, Gokcen Eren, Feyyaz Mıhoğlugil, Demet Akalgan, Mahmut Miski. Butyrylcholinesterase-inhibiting natural coumarin molecules as potential leads. Phytochemistry Letters 2021, 44 , 48-54. https://doi.org/10.1016/j.phytol.2021.05.001
    98. Flávia B. Lopes, Cecília M. S. Q. Aranha, João Paulo S. Fernandes. Histamine H 3 receptor and cholinesterases as synergistic targets for cognitive decline: Strategies to the rational design of multitarget ligands. Chemical Biology & Drug Design 2021, 98 (2) , 212-225. https://doi.org/10.1111/cbdd.13866
    99. Abdullah Al Mamun, Filip Pidaný, Daniela Hulcová, Jana Maříková, Tomáš Kučera, Monika Schmidt, Maria Carmen Catapano, Martina Hrabinová, Daniel Jun, Lubica Múčková, Jiří Kuneš, Jiří Janoušek, Rudolf Andrýs, Lucie Nováková, Rozálie Peřinová, Negar Maafi, Ondřej Soukup, Jan Korábečný, Lucie Cahlíková. Amaryllidaceae Alkaloids of Norbelladine-Type as Inspiration for Development of Highly Selective Butyrylcholinesterase Inhibitors: Synthesis, Biological Activity Evaluation, and Docking Studies. International Journal of Molecular Sciences 2021, 22 (15) , 8308. https://doi.org/10.3390/ijms22158308
    100. Fatima Y. Adeowo, Murtala A. Ejalonibu, Ahmed A. Elrashedy, Monsurat M. Lawal, Hezekiel M. Kumalo. Multi-target approach for Alzheimer’s disease treatment: computational biomolecular modeling of cholinesterase enzymes with a novel 4- N -phenylaminoquinoline derivative reveal promising potentials. Journal of Biomolecular Structure and Dynamics 2021, 39 (11) , 3825-3841. https://doi.org/10.1080/07391102.2020.1826129
    Load more citations

    Journal of Medicinal Chemistry

    Cite this: J. Med. Chem. 2014, 57, 19, 8167–8179
    Click to copy citationCitation copied!
    https://doi.org/10.1021/jm501195e
    Published September 16, 2014
    Copyright © 2014 American Chemical Society

    Article Views

    3965

    Altmetric

    -

    Citations

    Learn about these metrics

    Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

    Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

    The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.