1
|
Bernard PJ, Bellili D, Ismaili L. Calcium channel blockers' contribution to overcoming Current drug discovery challenges in Alzheimer's disease. Expert Opin Drug Discov 2024; 19:21-32. [PMID: 37800853 DOI: 10.1080/17460441.2023.2266994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive, irreversible, and multifactorial brain disorder that gradually and insidiously destroys individual's memory, thinking, and other cognitive abilities. AREAS COVERED In this perspective, the authors examine the complex and multifactorial nature of Alzheimer's disease and believe that the best approach to develop new drugs is the MTDL strategy, which obviously faces several challenges. These challenges include identifying the key combination of targets and their suitability for coordinated actions, as well as developing an acceptable pharmacokinetic and toxicological profile to deliver a drug candidate. EXPERT OPINION Since calcium plays a crucial role in the pathology of AD, a polypharmacological approach with calcium channel blockers reinforced by activities targeting other factors involved in AD is a serious option in our opinion. This is exemplified by a phase III clinical trial using a drug combination approach with Losartan, Amlodipine (a calcium channel blocker), and Atorvastatin, as well as several MTDL-based calcium channel blockade approaches with a promising in vitro and in vivo profile.
Collapse
Affiliation(s)
- Paul J Bernard
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| | - Djamila Bellili
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| | - Lhassane Ismaili
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| |
Collapse
|
2
|
Madar P, Nagalapur P, Chaudhari S, Sharma D, Koparde A, Buchade R, Kshirsagar S, Uttekar P, Jadhav S, Chaudhari P. The Unveiling of Therapeutic Targets for Alzheimer's Disease: An Integrative Review. Curr Top Med Chem 2024; 24:850-868. [PMID: 38424435 DOI: 10.2174/0115680266282492240220101049] [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: 10/23/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
Alzheimer's disease (AD) is characterized by a complex pathological landscape, necessitating a comprehensive treatment approach. This concise review paper delves into the idea of addressing multiple mechanisms in AD, summarizing the latest research findings on pathogenesis, risk factors, diagnostics, and therapeutic strategies. The etiology of AD is multifaceted, involving genetic, environmental, and lifestyle factors. The primary feature is the accumulation of amyloid-- beta and tau proteins, leading to neuroinflammation, synaptic dysfunction, oxidative stress, and neuronal loss. Conventional single-target therapies have shown limited effectiveness, prompting a shift toward simultaneously addressing multiple disease-related processes. Recent advancements in AD research underscore the potential of multifaceted therapies. This review explores strategies targeting both tau aggregation and amyloid-beta, along with interventions to alleviate neuroinflammation, enhance synaptic function, and reduce oxidative stress. In conclusion, the review emphasizes the growing importance of addressing various pathways in AD treatment. A holistic approach that targets different aspects of the disease holds promise for developing effective treatments and improving the quality of life for Alzheimer's patients and their caregivers.
Collapse
Affiliation(s)
- Pratiksha Madar
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Pooja Nagalapur
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Somdatta Chaudhari
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Devesh Sharma
- Department of Biotechnology, National JALMA Institute for Leprosy & Other Mycobacterial Diseases, Agra, India
| | - Akshada Koparde
- Department of Pharmaceutical Chemistry, Krishna Foundation's Jaywant Institute of Pharmacy, Malkapur, Karad, India
| | - Rahul Buchade
- Department of Pharmaceutical Chemistry, Indira College of Pharmacy, Tathwade, Pune, India
| | - Sandip Kshirsagar
- Department of Pharmaceutical Chemistry, Dr. D Y Patil College of Pharmacy, Pune, India
| | - Pravin Uttekar
- Department of Pharmacuetics, Savitribai Phule Pune University, Pune, India
| | - Shailaja Jadhav
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Praveen Chaudhari
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
3
|
Zaręba P, Łątka K, Mazur G, Gryzło B, Pasieka A, Godyń J, Panek D, Skrzypczak-Wiercioch A, Höfner GC, Latacz G, Maj M, Espargaró A, Sabaté R, Jóźwiak K, Wanner KT, Sałat K, Malawska B, Kulig K, Bajda M. Discovery of novel multifunctional ligands targeting GABA transporters, butyrylcholinesterase, β-secretase, and amyloid β aggregation as potential treatment of Alzheimer's disease. Eur J Med Chem 2023; 261:115832. [PMID: 37837674 DOI: 10.1016/j.ejmech.2023.115832] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/12/2023] [Accepted: 09/23/2023] [Indexed: 10/16/2023]
Abstract
Alzheimer's disease (AD) is a global health problem in the medical sector that will increase over time. The limited treatment of AD leads to the search for a new clinical candidate. Considering the multifactorial nature of AD, a strategy targeting number of regulatory proteins involved in the development of the disease is an effective approach. Here, we present a discovery of new multi-target-directed ligands (MTDLs), purposely designed as GABA transporter (GAT) inhibitors, that successfully provide the inhibitory activity against butyrylcholinesterase (BuChE), β-secretase (BACE1), amyloid β aggregation and calcium channel blockade activity. The selected GAT inhibitors, 19c and 22a - N-benzylamide derivatives of 4-aminobutyric acid, displayed the most prominent multifunctional profile. Compound 19c (mGAT1 IC50 = 10 μM, mGAT4 IC50 = 12 μM and BuChE IC50 = 559 nM) possessed the highest hBACE1 and Aβ40 aggregation inhibitory activity (IC50 = 1.57 μM and 99 % at 10 μM, respectively). Additionally, it showed a decrease in both the elongation and nucleation constants of the amyloid aggregation process. In contrast compound 22a represented the highest activity and a mixed-type of eqBuChE inhibition (IC50 = 173 nM) with hBACE1 (IC50 = 9.42 μM), Aβ aggregation (79 % at 10 μM) and mGATs (mGAT1 IC50 = 30 μM, mGAT4 IC50 = 25 μM) inhibitory activity. Performed molecular docking studies described the mode of interactions with GATs and enzymatic targets. In ADMET in vitro studies both compounds showed acceptable metabolic stability and low neurotoxicity. Successfully, compounds 19c and 22a at the dose of 30 mg/kg possessed statistically significant antiamnesic properties in a mouse model of amnesia caused by scopolamine and assessed in the novel object recognition (NOR) task or the passive avoidance (PA) task.
Collapse
Affiliation(s)
- Paula Zaręba
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Kamil Łątka
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Gabriela Mazur
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Beata Gryzło
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Anna Pasieka
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Justyna Godyń
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Dawid Panek
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Anna Skrzypczak-Wiercioch
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Kraków, Mickiewicz 24/28 St., 30-059, Kraków, Poland
| | - Georg C Höfner
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München Butenandtstr., 5-13, 81377, Munich, Germany
| | - Gniewomir Latacz
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, W. Chodzki 4a St., 20-093, Lublin, Poland
| | - Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028, Barcelona, Spain
| | - Raimon Sabaté
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), Av Joan XXIII, S/N, 08028, Barcelona, Spain
| | - Krzysztof Jóźwiak
- Department of Biopharmacy, Medical University of Lublin, W. Chodzki 4a St., 20-093, Lublin, Poland
| | - Klaus T Wanner
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München Butenandtstr., 5-13, 81377, Munich, Germany
| | - Kinga Sałat
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Barbara Malawska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Katarzyna Kulig
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland
| | - Marek Bajda
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 St., 30-688, Kraków, Poland.
| |
Collapse
|
4
|
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: 5.0] [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
|
5
|
Novel indolotacrine hybrids as acetylcholinesterase inhibitors: design, synthesis, biological evaluation, and molecular docking studies. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2023. [DOI: 10.1007/s13738-022-02726-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
6
|
Djafarou S, Amine Khodja I, Boulebd H. Computational design of new tacrine analogs: an in silico prediction of their cholinesterase inhibitory, antioxidant, and hepatotoxic activities. J Biomol Struct Dyn 2023; 41:91-105. [PMID: 34825629 DOI: 10.1080/07391102.2021.2004232] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tacrine, the first drug approved for the treatment of Alzheimer's disease (AD), is a non-competitive cholinesterase inhibitor withdrawn due to its acute hepatotoxicity. However, new non-hepatotoxic forms of tacrine have been actively researched. Moreover, several recent reports have shown that oxidative stress is the cause of damage and plays a role in the pathogenesis of several neurodegenerative diseases including AD. The aim of the present study is the design of new easily synthesized tacrine analogs with less hepatotoxicity and potent antioxidant activity. In this context, a library of 34 novel tacrine analogs bearing an antioxidant fragment was designed and evaluated for its hepatotoxicity as well as anticholinesterase and antioxidant activities using computational methods. As a result, six new tacrine analogs have been proposed as potential inhibitors of cholinesterase with antioxidant activity and low or no hepatotoxicity. Furthermore, ADME calculations suggest that these compounds are promising oral drug candidates. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Selsabil Djafarou
- Laboratory of Synthesis of Molecules with Biological Interest, University of Frères Mentouri Constantine 1, Constantine, Algeria
| | - Imene Amine Khodja
- Laboratory of Synthesis of Molecules with Biological Interest, University of Frères Mentouri Constantine 1, Constantine, Algeria
| | - Houssem Boulebd
- Laboratory of Synthesis of Molecules with Biological Interest, University of Frères Mentouri Constantine 1, Constantine, Algeria
| |
Collapse
|
7
|
Malek R, Simakov A, Davis A, Maj M, Bernard PJ, Wnorowski A, Martin H, Marco-Contelles J, Chabchoub F, Dallemagne P, Rochais C, Jozwiak K, Ismaili L. Biginelli Reaction Synthesis of Novel Multitarget-Directed Ligands with Ca 2+ Channel Blocking Ability, Cholinesterase Inhibition, Antioxidant Capacity, and Nrf2 Activation. Molecules 2022; 28:molecules28010071. [PMID: 36615267 PMCID: PMC9822022 DOI: 10.3390/molecules28010071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Novel multitarget-directed ligands BIGI 4a-d and BIGI 5a-d were designed and synthesized with a simple and cost-efficient procedure via a one-pot three-component Biginelli reaction targeting acetyl-/butyrylcholinesterases inhibition, calcium channel antagonism, and antioxidant ability. Among these multitarget-directed ligands, BIGI 4b, BIGI 4d, and BIGI 5b were identified as promising new hit compounds showing in vitro balanced activities toward the recognized AD targets. In addition, these compounds showed suitable physicochemical properties and a good druglikeness score predicted by Data Warrior software.
Collapse
Affiliation(s)
- Rim Malek
- Laboratoire LINC UR 481, Pôle de Chimie Médicinale, University Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France
- Laboratory of Applied Chemistry: Heterocycles, Lipids and Polymers, Faculty of Sciences of Sfax, University of Sfax, Sfax 3000, Tunisia
| | - Alexey Simakov
- PEPITE EA4267, University Franche-Comté, F-25000 Besançon, France
| | - Audrey Davis
- Centre d’Etudes et de Recherche sur le Médicament de Normandie, Normandie University, Unicaen, CERMN, 14000 Caen, France
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland
| | - Paul J. Bernard
- Laboratoire LINC UR 481, Pôle de Chimie Médicinale, University Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France
| | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland
| | - Helene Martin
- PEPITE EA4267, University Franche-Comté, F-25000 Besançon, France
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC), C/ Juan de la Cierva 3, 28006 Madrid, Spain
| | - Fakher Chabchoub
- Laboratory of Applied Chemistry: Heterocycles, Lipids and Polymers, Faculty of Sciences of Sfax, University of Sfax, Sfax 3000, Tunisia
| | - Patrick Dallemagne
- Centre d’Etudes et de Recherche sur le Médicament de Normandie, Normandie University, Unicaen, CERMN, 14000 Caen, France
| | - Christophe Rochais
- Centre d’Etudes et de Recherche sur le Médicament de Normandie, Normandie University, Unicaen, CERMN, 14000 Caen, France
| | - Krzysztof Jozwiak
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland
| | - Lhassane Ismaili
- Laboratoire LINC UR 481, Pôle de Chimie Médicinale, University Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France
- Correspondence:
| |
Collapse
|
8
|
Multicomponent reactions as a privileged tool for multitarget-directed ligand strategies in Alzheimer's disease therapy. Future Med Chem 2022; 14:1583-1606. [PMID: 36263996 DOI: 10.4155/fmc-2022-0170] [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: 11/17/2022] Open
Abstract
Among neurodegenerative pathologies affecting the older population, Alzheimer's disease is the most common type of dementia and leads to neurocognitive and behavioral disorders. It is a complex and progressive age-related multifactorial disease characterized by a series of highly interconnected pathophysiological processes. Within the last decade, the multitarget-directed ligand strategy has emerged as a viable approach to developing complex molecules that exhibit several pharmacophores which can target the different enzymes and receptors involved in the pathogenesis of the disease. Herein, we focus on using multicomponent reactions such as Hantzsch, Biginelli and Ugi to develop these biologically active multitopic ligands.
Collapse
|
9
|
Mitra S, Muni M, Shawon NJ, Das R, Emran TB, Sharma R, Chandran D, Islam F, Hossain MJ, Safi SZ, Sweilam SH. Tacrine Derivatives in Neurological Disorders: Focus on Molecular Mechanisms and Neurotherapeutic Potential. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7252882. [PMID: 36035218 PMCID: PMC9410840 DOI: 10.1155/2022/7252882] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/19/2022] [Accepted: 08/03/2022] [Indexed: 12/13/2022]
Abstract
Tacrine is a drug used in the treatment of Alzheimer's disease as a cognitive enhancer and inhibitor of the enzyme acetylcholinesterase (AChE). However, its clinical application has been restricted due to its poor therapeutic efficacy and high prevalence of detrimental effects. An attempt was made to understand the molecular mechanisms that underlie tacrine and its analogues influence over neurotherapeutic activity by focusing on modulation of neurogenesis, neuroinflammation, endoplasmic reticulum stress, apoptosis, and regulatory role in gene and protein expression, energy metabolism, Ca2+ homeostasis modulation, and osmotic regulation. Regardless of this, analogues of tacrine are considered as a model inhibitor of cholinesterase in the therapy of Alzheimer's disease. The variety both in structural make-up and biological functions of these substances is the main appeal for researchers' interest in them. A new paradigm for treating neurological diseases is presented in this review, which includes treatment strategies for Alzheimer's disease, as well as other neurological disorders like Parkinson's disease and the synthesis and biological properties of newly identified versatile tacrine analogues and hybrids. We have also shown that these analogues may have therapeutic promise in the treatment of neurological diseases in a variety of experimental systems.
Collapse
Affiliation(s)
- Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Maniza Muni
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Nusrat Jahan Shawon
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 Uttar Pradesh, India
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, Tamil Nadu 642109, India
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Md. Jamal Hossain
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road, Dhanmondi, Dhaka 1205, Bangladesh
| | - Sher Zaman Safi
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, 42610 Selangor, Malaysia
- IRCBM, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City 11829, Egypt
| |
Collapse
|
10
|
Alzheimer's disease: Updated multi-targets therapeutics are in clinical and in progress. Eur J Med Chem 2022; 238:114464. [DOI: 10.1016/j.ejmech.2022.114464] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
|
11
|
Chehardoli G, Gholamhoseini P, Ebadi A, Ziaei M, Akbarzadeh T, Saeedi M, Mahdavi M, Khoshneviszadeh M, Najafi Z. 6‐Methoxy‐1‐tetralone Derivatives Bearing an N‐Arylpyridinium Moiety as Cholinesterase Inhibitors: Design, Synthesis, Biological Evaluation, and Molecular Docking Study. ChemistrySelect 2022. [DOI: 10.1002/slct.202201977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Gholamabbas Chehardoli
- Department of Medicinal Chemistry School of Pharmacy Medicinal Plants and Natural Products Research Center Hamadan University of Medical Sciences Shahid Fahmideh Street 6517838678 Hamadan Iran
| | - Pooriya Gholamhoseini
- Department of Medicinal Chemistry School of Pharmacy Hamadan University of Medical Sciences Shahid Fahmideh Street 6517838678 Hamadan Iran
| | - Ahmad Ebadi
- Department of Medicinal Chemistry School of Pharmacy Medicinal Plants and Natural Products Research Center Hamadan University of Medical Sciences Shahid Fahmideh Street 6517838678 Hamadan Iran
| | - Maral Ziaei
- Department of Medicinal Chemistry School of Pharmacy Hamadan University of Medical Sciences Shahid Fahmideh Street 6517838678 Hamadan Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry Faculty of Pharmacy Tehran University of Medical Sciences 16 Azar Street 1417614411 Tehran Iran
- Persian Medicine and Pharmacy Research Center Tehran University of Medical Sciences 16 Azar Street 1417614411 Tehran Iran
| | - Mina Saeedi
- Persian Medicine and Pharmacy Research Center Tehran University of Medical Sciences 16 Azar Street 1417614411 Tehran Iran
- Medicinal Plants Research Center, Faculty of Pharmacy Tehran University of Medical Sciences 16 Azar Street 1417614411 Tehran Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences 1411713137 Tehran Iran
| | - Mehdi Khoshneviszadeh
- Department of Medicinal Chemistry Faculty of Pharmacy Shiraz University of Medical Sciences 7146864685 Shiraz Iran
| | - Zahra Najafi
- Department of Medicinal Chemistry School of Pharmacy Hamadan University of Medical Sciences Shahid Fahmideh Street 6517838678 Hamadan Iran
| |
Collapse
|
12
|
Effects of Linkers and Substitutions on Multitarget Directed Ligands for Alzheimer’s Diseases: Emerging Paradigms and Strategies. Int J Mol Sci 2022; 23:ijms23116085. [PMID: 35682763 PMCID: PMC9181730 DOI: 10.3390/ijms23116085] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is multifactorial, progressive and the most predominant cause of cognitive impairment and dementia worldwide. The current “one-drug, one-target” approach provides only symptomatic relief to the condition but is unable to cure the disease completely. The conventional single-target therapeutic approach might not always induce the desired effect due to the multifactorial nature of AD. Hence, multitarget strategies have been proposed to simultaneously knock out multiple targets involved in the development of AD. Herein, we provide an overview of the various strategies, followed by the multitarget-directed ligand (MTDL) development, rationale designs and efficient examples. Furthermore, the effects of the linkers and substitutional functional groups on MTDLs against various targets of AD and their modes of action are also discussed.
Collapse
|
13
|
Moreira NCDS, Lima JEBDF, Marchiori MF, Carvalho I, Sakamoto-Hojo ET. Neuroprotective Effects of Cholinesterase Inhibitors: Current Scenario in Therapies for Alzheimer's Disease and Future Perspectives. J Alzheimers Dis Rep 2022; 6:177-193. [PMID: 35591949 PMCID: PMC9108627 DOI: 10.3233/adr-210061] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a slowly progressive neurodegenerative disease conceptualized as a continuous process, ranging from mild cognitive impairment (MCI), to the mild, moderate, and severe clinical stages of AD dementia. AD is considered a complex multifactorial disease. Currently, the use of cholinesterase inhibitors (ChEI), such as tacrine, donepezil, rivastigmine, and galantamine, has been the main treatment for AD patients. Interestingly, there is evidence that ChEI also promotes neuroprotective effects, bringing some benefits to AD patients. The mechanisms by which the ChEI act have been investigated in AD. ChEI can modulate the PI3K/AKT pathway, which is an important signaling cascade that is capable of causing a significant functional impact on neurons by activating cell survival pathways to promote neuroprotective effects. However, there is still a huge challenge in the field of neuroprotection, but in the context of unravelling the details of the PI3K/AKT pathway, a new scenario has emerged for the development of more efficient drugs that act on multiple protein targets. Thus, the mechanisms by which ChEI can promote neuroprotective effects and prospects for the development of new drug candidates for the treatment of AD are discussed in this review.
Collapse
Affiliation(s)
| | | | - Marcelo Fiori Marchiori
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
14
|
Przybyłowska M, Dzierzbicka K, Kowalski S, Demkowicz S, Daśko M, Inkielewicz-Stepniak I. Design, synthesis and biological evaluation of novel N-phosphorylated and O-phosphorylated tacrine derivatives as potential drugs against Alzheimer's disease. J Enzyme Inhib Med Chem 2022; 37:1012-1022. [PMID: 35361039 PMCID: PMC8979514 DOI: 10.1080/14756366.2022.2045591] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
In this work, we designed, synthesised and biologically investigated a novel series of 14 N- and O-phosphorylated tacrine derivatives as potential anti-Alzheimer’s disease agents. In the reaction of 9-chlorotacrine and corresponding diamines/aminoalkylalcohol we obtained diamino and aminoalkylhydroxy tacrine derivatives. Next, the compounds were acid to give final products 6–13 and 16–21 that were characterised by 1H, 13 C, 31 P NMR and MS. The results of the docking studies revealed that the designed phosphorus hybrids, in theory can bind to AChE and BChE. All compounds exhibited significantly lower AutoDock Vina scores compared to tacrine. The inhibitory potency evaluation was performed using the Ellman’s method. The most inhibitory activity against AChE exhibited compound 8 with an IC50 value of 6.11 nM and against BChE 13 with an IC50 value of 1.97 nM and they were 6- and 12-fold potent than tacrine. Compound 19 showed the lack of hepatocytotoxicity in MTT assay.
Collapse
Affiliation(s)
- Maja Przybyłowska
- Department of Organic Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Szymon Kowalski
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Mateusz Daśko
- Department of Inorganic Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
15
|
Javed MA, Ashraf N, Saeed Jan M, Mahnashi MH, Alqahtani YS, Alyami BA, Alqarni AO, Asiri YI, Ikram M, Sadiq A, Rashid U. Structural Modification, In Vitro, In Vivo, Ex Vivo, and In Silico Exploration of Pyrimidine and Pyrrolidine Cores for Targeting Enzymes Associated with Neuroinflammation and Cholinergic Deficit in Alzheimer's Disease. ACS Chem Neurosci 2021; 12:4123-4143. [PMID: 34643082 DOI: 10.1021/acschemneuro.1c00507] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
To obtain a multipotent framework that can target simultaneously COX-2, 5-LOX, acetylcholinesterase (AChE), and butyrylcholinesterase (BChE) to treat neuroinflammation, a series of derivatives containing pyrimidine and pyrrolidine cores were rationally synthesized and evaluated. Pyrazoline-pyrimidine hybrid (23g), (3-acetylcoumarin derivative of pyrrolidin-1-yl)benzenesulfonamide (27), and tacrine derivatives of (pyrrolidin-1-yl)benzenesulfonamide (31, 38) displayed excellent in vitro COX-2 inhibition having IC50 value in the nanomolar range. Tacrine-pyrrolidine hybrids 36 and 38, and tacrine-pyrimidine hybrid (46) emerged as the most potent eeAChE inhibitors with IC50 values of 23, 16, and 2 nM, respectively. However, compounds 27, 31, and 38 possessed excellent simultaneous and balanced inhibitory activity against all of the four tested targets and thus emerged as optimal multipotent hybrid compounds among all of the synthesized series of the compounds. In the ex vivo, transgenic animal models treated with compounds 36 and 46 displayed a significant decline in both AChE and BChE potentials in the hippocampus and cortical tissues. In anti-inflammatory activities, animals treated with compounds 36 and 46 displayed a significant % inhibition of edema induced by carrageenan and arachidonic acid. Biochemical analysis and histopathological examination of mice liver indicate that tacrine derivatives are devoid of hepatotoxicity and neurotoxicity against SH-SY5Y neuroblastoma cell lines. In vivo acute toxicity study showed the safety of synthesized compounds up to 1000 mg/kg dose. The inhibitory manner of interaction of these potent drugs on all of the studied in vitro targets was confirmed by molecular docking investigations.
Collapse
Affiliation(s)
- Muhammad Aamir Javed
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| | - Nighat Ashraf
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| | | | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Yahya S. Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Bandar A. Alyami
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Ali O. Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, 1988 Najran, Saudi Arabia
| | - Yahya I. Asiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, 1882 Abha, Saudi Arabia
| | - Muhammad Ikram
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, KP, Pakistan
| | - Abdul Sadiq
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, 18000 Chakdara, Dir (L), KP, Pakistan
| | - Umer Rashid
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060 Abbottabad, Pakistan
| |
Collapse
|
16
|
Sadafi Kohnehshahri M, Chehardoli G, Bahiraei M, Akbarzadeh T, Ranjbar A, Rastegari A, Najafi Z. Novel tacrine-based acetylcholinesterase inhibitors as potential agents for the treatment of Alzheimer's disease: Quinolotacrine hybrids. Mol Divers 2021; 26:489-503. [PMID: 34491490 DOI: 10.1007/s11030-021-10307-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/26/2021] [Indexed: 11/28/2022]
Abstract
A new series of quinolotacrine hybrids including cyclopenta- and cyclohexa-quinolotacrine derivatives were designed, synthesized, and assessed as anti-cholinesterase (ChE) agents. The designed derivatives indicated higher inhibitory effect on the acetylcholinesterase (AChE) with IC50 values of 0.285-100 µM compared to butyrylcholinesterase (BChE) with IC50 values of > 100 µM. Of these compounds, cyclohexa-quinolotacrine hybrids displayed a little better anti-AChE activity than cyclopenta-quinolotacrine hybrids. Compound 8-amino-7-(3-hydroxyphenyl)-5,7,9,10,11,12-hexahydro-6H-pyrano[2,3-b:5,6-c'] diquinolin-6-one (6m) including 3-hydroxyphenyl and cyclohexane ring moieties exhibited the best AChE inhibitory activity with IC50 value of 0.285 µM. The kinetic and molecular docking studies indicated that compound 6m occupied both the catalytic anionic site (CAS) and peripheral anionic site (PAS) of AChE as a mixed inhibitor. Using neuroprotective assay against H2O2-induced cell death in PC12 cells, the compound 6h illustrated significant protection among the assessed compounds. In silico ADME studies estimated good drug-likeness for the designed compounds. As a result, these quinolotacrine hybrids can be very encouraging AChE inhibitors to treat Alzheimer's disease. A novel series of quinolotacrine hybrids were designed, synthesized, and evaluated against AChE and BChE enzymes as potential agents for the treatment of AD. The hybrids showed good to significant inhibitory activity against AChE (0.285-100 μM) compared to butyrylcholinesterase (BChE) with IC50 values of > 100 μM. Among them, compound 8-amino-7-(3-hydroxyphenyl)-5,7,9,10,11,12-hexahydro-6H-pyrano[2,3-b:5,6-c'] diquinolin-6-one (6 m) bearing 3-hydroxyphenyl moiety and cyclohexane ring exhibited the highest anti-AChE activity with IC50 value of 0.285 μM. The kinetic and molecular docking studies illustrated that compound 6 m is a mixed inhibitor and binds to both the catalytic anionic site (CAS) and peripheral anionic site (PAS) of AChE.
Collapse
Affiliation(s)
- Mehrdad Sadafi Kohnehshahri
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gholamabbas Chehardoli
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoomeh Bahiraei
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Ranjbar
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arezoo Rastegari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Najafi
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
17
|
Tabassum R, Ashfaq M, Oku H. Current Pharmaceutical Aspects of Synthetic Quinoline Derivatives. Mini Rev Med Chem 2021; 21:1152-1172. [PMID: 33319670 DOI: 10.2174/1389557520999201214234735] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022]
Abstract
Quinoline derivatives are considered broad-spectrum pharmacological compounds that exhibit a wide range of biological activities. Integration of quinoline moiety can improve its physical and chemical properties and also pharmacological behavior. Due to its wide range of pharmaceutical applications, it is a very popular compound to design new drugs for the treatment of multiple diseases like cancer, dengue fever, malaria, tuberculosis, fungal infections, AIDS, Alzheimer's disease and diabetes. In this review, our major focus is to pay attention to the biological activities of quinoline compounds in the treatment of these diseases such as anti-viral, anti-cancer, anti-malarial, antibacterial, anti-fungal, anti-tubercular and anti-diabetic.
Collapse
Affiliation(s)
- Rukhsana Tabassum
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, 36100, Pakistan
| | - Muhammad Ashfaq
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, 36100, Pakistan
| | - Hiroyuki Oku
- Division of Molecular Science, Graduate School of Science & Engineering Gunma University, Gunma 376-8515, Japan
| |
Collapse
|
18
|
Babaee S, Chehardoli G, Akbarzadeh T, Zolfigol MA, Mahdavi M, Rastegari A, Homayouni Moghadam F, Najafi Z. Design, Synthesis, and Molecular Docking of Some Novel Tacrine Based Cyclopentapyranopyridine- and Tetrahydropyranoquinoline-Kojic Acid Derivatives as Anti-Acetylcholinesterase Agents. Chem Biodivers 2021; 18:e2000924. [PMID: 33861892 DOI: 10.1002/cbdv.202000924] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/15/2021] [Indexed: 12/13/2022]
Abstract
A novel series of tacrine based cyclopentapyranopyridine- and tetrahydropyranoquinoline-kojic acid derivatives were designed, synthesized, and evaluated as anti-cholinesterase agents. The chemical structures of all target compounds were characterized by 1 H-NMR, 13 C-NMR, and elemental analyses. The synthesized compounds mostly inhibited acetylcholinesterase enzyme (AChE) with IC50 values of 4.18-48.71 μM rather than butyrylcholinesterase enzyme (BChE) with IC50 values of >100 μM. Among them, cyclopentapyranopyridine-kojic acid derivatives showed slightly better AChE inhibitory activity compared to tetrahydropyranoquinoline-kojic acid. The compound 10-amino-2-(hydroxymethyl)-11-(4-isopropylphenyl)-7,8,9,11-tetrahydro-4H-cyclopenta[b]pyrano[2',3' : 5,6]pyrano[3,2-e]pyridin-4-one (6f) bearing 4-isopropylphenyl moiety and cyclopentane ring exhibited the highest anti-AChE activity with IC50 value of 4.18 μM. The kinetic study indicated that the compound 6f acts as a mixed inhibitor and the molecular docking studies also illustrated that the compound 6f binds to both the catalytic site (CS) and peripheral anionic site (PAS) of AChE. The compound 6f showed moderate neuroprotective properties against H2 O2 -induced cytotoxicity in PC12 cells. The theoretical ADME study also predicted good drug-likeness for the compound 6f. Based on these results, the compound 6f seems to be a very promising AChE inhibitor for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Saeed Babaee
- Department of Organic Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan, 6517838683, Iran
| | - Gholamabbas Chehardoli
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Mohammad Ali Zolfigol
- Department of Organic Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan, 6517838683, Iran
| | - Mohammad Mahdavi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, 8165131378, Iran
| | - Arezoo Rastegari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Farshad Homayouni Moghadam
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713137, Iran
| | - Zahra Najafi
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| |
Collapse
|
19
|
Wan LX, Zhen YQ, He ZX, Zhang Y, Zhang L, Li X, Gao F, Zhou XL. Late-Stage Modification of Medicine: Pd-Catalyzed Direct Synthesis and Biological Evaluation of N-Aryltacrine Derivatives. ACS OMEGA 2021; 6:9960-9972. [PMID: 33869976 PMCID: PMC8047743 DOI: 10.1021/acsomega.1c01404] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 05/13/2023]
Abstract
A new series of N-aryltacrine derivatives were designed and synthesized as cholinesterase inhibitors by the late-stage modification of tacrine, using the palladium-catalyzed Buchwald-Hartwig cross-coupling reaction. In vitro inhibition assay against acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) demonstrated that most of the synthesized compounds had potent AChE inhibitory activity with negative inhibition of BuChE. Among them, N-(4-(trifluoromethyl)phenyl)-tacrine (3g) and N-(4-methoxypyridin-2-yl)-tacrine (3o) showed the most potent activity against AChE (IC50 values of 1.77 and 1.48 μM, respectively). The anti-AChE activity of 3g and 3o was 3.5 times more than that of tacrine (IC50 value of 5.16 μM). Compound 3o also displayed anti-BuChE activity with an IC50 value of 19.00 μM. Cell-based assays against HepG2 and SH-SY5Y cell lines revealed that 3o had significantly lower hepatotoxicity compared to tacrine, with additional neuroprotective activity against H2O2-induced damage in SH-SY5Y cells. The advantages including synthetic accessibility, high potency, low toxicity, and adjunctive neuroprotective activity make compound 3o a new promising multifunctional candidate for the treatment of Alzheimer's disease.
Collapse
|
20
|
Pachón Angona I, Martin H, Daniel S, Moraleda I, Bonet A, Wnorowski A, Maj M, Jozwiak K, Iriepa I, Refouvelet B, Marco-Contelles J, Ismaili L. Synthesis of Hantzsch Adducts as Cholinesterases and Calcium Flux inhibitors, Antioxidants and Neuroprotectives. Int J Mol Sci 2020; 21:ijms21207652. [PMID: 33081112 PMCID: PMC7589057 DOI: 10.3390/ijms21207652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 01/22/2023] Open
Abstract
We report herein the design, synthesis, biological evaluation, and molecular modelling of new inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), able to block Ca+2 channels also showing antioxidant and neuroprotective activities. The new MTDL, dialkyl 2,6-dimethyl-4-(4-((5-aminoalkyl)oxy)phenyl)-1,4-dihydropyridine-3,5-dicarboxylate 3a-p, have been obtained via Hantzsch reaction from appropriate and commercially available precursors. Pertinent biological analysis has prompted us to identify MTDL 3h [dimethyl-4-(4-((5-(4-benzylpiperidin-1-yl)pentyl)oxy)phenyl)-2,6-dimethyl-1,4-dihydropyridine-3,5-dicarboxylate] as an attractive inhibitor of AChE (1.8 μM) and BuChE (2 μM), Ca+2 channel antagonist (47.72% at 10 μM), and antioxidant (2.54 TE) agent, showing significant neuroprotection 28.68% and 38.29% against H2O2, and O/R, respectively, at 0.3 μM, thus being considered a hit-compound for further investigation in our search for anti-Alzheimer's disease agents.
Collapse
Affiliation(s)
- Irene Pachón Angona
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
| | - Helene Martin
- PEPITE EA4267, Laboratoire de Toxicologie Cellulaire, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France; (H.M.); (A.B.)
| | - Solene Daniel
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
| | - Ignacio Moraleda
- Department of Organic Chemistry and Inorganic Chemistry, School Sciences, University of Alcalá, Ctra. Barcelona, Km. 33.6, 28871 Alcalá de Henares, Spain;
| | - Alexandre Bonet
- PEPITE EA4267, Laboratoire de Toxicologie Cellulaire, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France; (H.M.); (A.B.)
| | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Krzysztof Jozwiak
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Isabel Iriepa
- Department of Organic Chemistry and Inorganic Chemistry, School Sciences, University of Alcalá, Ctra. Barcelona, Km. 33.6, 28871 Alcalá de Henares, Spain;
- Correspondence: (I.I.); (B.R.); (L.I.)
| | - Bernard Refouvelet
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
- Correspondence: (I.I.); (B.R.); (L.I.)
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC), Juan de la Cierva, 3, 28006-Madrid, Spain;
| | - Lhassane Ismaili
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.)
- Correspondence: (I.I.); (B.R.); (L.I.)
| |
Collapse
|
21
|
Merged Tacrine-Based, Multitarget-Directed Acetylcholinesterase Inhibitors 2015-Present: Synthesis and Biological Activity. Int J Mol Sci 2020; 21:ijms21175965. [PMID: 32825138 PMCID: PMC7504404 DOI: 10.3390/ijms21175965] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 11/17/2022] Open
Abstract
Acetylcholinesterase is an important biochemical enzyme in that it controls acetylcholine-mediated neuronal transmission in the central nervous system, contains a unique structure with two binding sites connected by a gorge region, and it has historically been the main pharmacological target for treatment of Alzheimer's disease. Given the large projected increase in Alzheimer's disease cases in the coming decades and its complex, multifactorial nature, new drugs that target multiple aspects of the disease at once are needed. Tacrine, the first acetylcholinesterase inhibitor used clinically but withdrawn due to hepatotoxicity concerns, remains an important starting point in research for the development of multitarget-directed acetylcholinesterase inhibitors. This review highlights tacrine-based, multitarget-directed acetylcholinesterase inhibitors published in the literature since 2015 with a specific focus on merged compounds (i.e., compounds where tacrine and a second pharmacophore show significant overlap in structure). The synthesis of these compounds from readily available starting materials is discussed, along with acetylcholinesterase inhibition data, relative to tacrine, and structure activity relationships. Where applicable, molecular modeling, to elucidate key enzyme-inhibitor interactions, and secondary biological activity is highlighted. Of the numerous compounds identified, there is a subset with promising preliminary screening results, which should inspire further development and future research in this field.
Collapse
|
22
|
Multitarget Therapeutic Strategies for Alzheimer's Disease: Review on Emerging Target Combinations. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5120230. [PMID: 32714977 PMCID: PMC7354643 DOI: 10.1155/2020/5120230] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases represent nowadays one of the major health problems. Despite the efforts made to unveil the mechanism leading to neurodegeneration, it is still not entirely clear what triggers this phenomenon and what allows its progression. Nevertheless, it is accepted that neurodegeneration is a consequence of several detrimental processes, such as protein aggregation, oxidative stress, and neuroinflammation, finally resulting in the loss of neuronal functions. Starting from these evidences, there has been a wide search for novel agents able to address more than a single event at the same time, the so-called multitarget-directed ligands (MTDLs). These compounds originated from the combination of different pharmacophoric elements which endowed them with the ability to interfere with different enzymatic and/or receptor systems, or to exert neuroprotective effects by modulating proteins and metal homeostasis. MTDLs have been the focus of the latest strategies to discover a new treatment for Alzheimer's disease (AD), which is considered the most common form of dementia characterized by neurodegeneration and cognitive dysfunctions. This review is aimed at collecting the latest and most interesting target combinations for the treatment of AD, with a detailed discussion on new agents with favorable in vitro properties and on optimized structures that have already been assessed in vivo in animal models of dementia.
Collapse
|
23
|
Pachòn Angona I, Daniel S, Martin H, Bonet A, Wnorowski A, Maj M, Jóźwiak K, Silva TB, Refouvelet B, Borges F, Marco-Contelles J, Ismaili L. Design, Synthesis and Biological Evaluation of New Antioxidant and Neuroprotective Multitarget Directed Ligands Able to Block Calcium Channels. Molecules 2020; 25:molecules25061329. [PMID: 32183349 PMCID: PMC7144121 DOI: 10.3390/molecules25061329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 01/08/2023] Open
Abstract
We report herein the design, synthesis and biological evaluation of new antioxidant and neuroprotective multitarget directed ligands (MTDLs) able to block Ca2+ channels. New dialkyl 2,6-dimethyl-4-(4-(prop-2-yn-1-yloxy)phenyl)-1,4-dihydropyridine-3,5-dicarboxylate MTDLs 3a–t, resulting from the juxtaposition of nimodipine, a Ca2+ channel antagonist, and rasagiline, a known MAO inhibitor, have been obtained from appropriate and commercially available precursors using a Hantzsch reaction. Pertinent biological analysis has prompted us to identify the MTDL 3,5-dimethyl-2,6–dimethyl–4-[4-(prop–2–yn–1-yloxy)phenyl]-1,4-dihydro- pyridine- 3,5-dicarboxylate (3a), as an attractive antioxidant (1.75 TE), Ca2+ channel antagonist (46.95% at 10 μM), showing significant neuroprotection (38%) against H2O2 at 10 μM, being considered thus a hit-compound for further investigation in our search for anti-Alzheimer’s disease agents.
Collapse
Affiliation(s)
- Irene Pachòn Angona
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.); (B.R.)
| | - Solene Daniel
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.); (B.R.)
| | - Helene Martin
- PEPITE EA4267, Laboratoire de Toxicologie Cellulaire, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France; (H.M.); (A.B.)
| | - Alexandre Bonet
- PEPITE EA4267, Laboratoire de Toxicologie Cellulaire, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France; (H.M.); (A.B.)
| | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Maciej Maj
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Krzysztof Jóźwiak
- Department of Biopharmacy, Medical University of Lublin, ul. W. Chodzki 4a, 20-093 Lublin, Poland; (A.W.); (M.M.); (K.J.)
| | - Tiago Barros Silva
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre 1021/1055, 4169-007 Porto, Portugal; (T.B.S.); (F.B.)
| | - Bernard Refouvelet
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.); (B.R.)
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre 1021/1055, 4169-007 Porto, Portugal; (T.B.S.); (F.B.)
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC), Juan de la Cierva, 3, 28006 Madrid, Spain
- Correspondence: (J.M.-C.); (L.I.)
| | - Lhassane Ismaili
- Neurosciences Intégratives et Cliniques EA 481, Pôle de Chimie Organique et Thérapeutique, Univ. Bourgogne Franche-Comté, UFR Santé, 19, rue Ambroise Paré, F-25000 Besançon, France; (I.P.A.); (S.D.); (B.R.)
- Correspondence: (J.M.-C.); (L.I.)
| |
Collapse
|
24
|
Xie J, Liang R, Wang Y, Huang J, Cao X, Niu B. Progress in Target Drug Molecules for Alzheimer's Disease. Curr Top Med Chem 2020; 20:4-36. [DOI: 10.2174/1568026619666191203113745] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/20/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease that 4 widespread in the elderly.
The etiology of AD is complicated, and its pathogenesis is still unclear. Although there are many
researches on anti-AD drugs, they are limited to reverse relief symptoms and cannot treat diseases.
Therefore, the development of high-efficiency anti-AD drugs with no side effects has become an urgent
need. Based on the published literature, this paper summarizes the main targets of AD and their drugs,
and focuses on the research and development progress of these drugs in recent years.
Collapse
Affiliation(s)
- Jiayang Xie
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Ruirui Liang
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Yajiang Wang
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Junyi Huang
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Xin Cao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai, China
| | - Bing Niu
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| |
Collapse
|
25
|
Sharma P, Tripathi MK, Shrivastava SK. Cholinesterase as a Target for Drug Development in Alzheimer's Disease. Methods Mol Biol 2020; 2089:257-286. [PMID: 31773661 DOI: 10.1007/978-1-0716-0163-1_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is an enormous healthcare challenge, and 50 million people are currently suffering from it. There are several pathophysiological mechanisms involved, but cholinesterase inhibitors remained the major target from the last 2-3 decades. Among four available therapeutics (donepezil, rivastigmine, galantamine, and memantine), three of them are cholinesterase inhibitors. Herein, we describe the role of acetylcholine sterase (AChE) and related hypothesis in AD along with the pharmacological and chemical aspects of the available cholinesterase inhibitors. This chapter discusses the development of several congeners and hybrids of available cholinesterase inhibitors along with their binding patterns in enzyme active sites.
Collapse
Affiliation(s)
- Piyoosh Sharma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Manish Kumar Tripathi
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushant Kumar Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India.
| |
Collapse
|
26
|
The chemistry toolbox of multitarget-directed ligands for Alzheimer's disease. Eur J Med Chem 2019; 181:111572. [DOI: 10.1016/j.ejmech.2019.111572] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 01/04/2023]
|
27
|
Malek R, Maj M, Wnorowski A, Jóźwiak K, Martin H, Iriepa I, Moraleda I, Chabchoub F, Marco-Contelles J, Ismaili L. Multi-target 1,4-dihydropyridines showing calcium channel blockade and antioxidant capacity for Alzheimer’s disease therapy. Bioorg Chem 2019; 91:103205. [DOI: 10.1016/j.bioorg.2019.103205] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/19/2019] [Accepted: 08/14/2019] [Indexed: 11/17/2022]
|
28
|
Pérez-Areales FJ, Turcu AL, Barniol-Xicota M, Pont C, Pivetta D, Espargaró A, Bartolini M, De Simone A, Andrisano V, Pérez B, Sabate R, Sureda FX, Vázquez S, Muñoz-Torrero D. A novel class of multitarget anti-Alzheimer benzohomoadamantane‒chlorotacrine hybrids modulating cholinesterases and glutamate NMDA receptors. Eur J Med Chem 2019; 180:613-626. [PMID: 31351393 DOI: 10.1016/j.ejmech.2019.07.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 10/26/2022]
Abstract
The development of multitarget compounds against multifactorial diseases, such as Alzheimer's disease, is an area of very intensive research, due to the expected superior therapeutic efficacy that should arise from the simultaneous modulation of several key targets of the complex pathological network. Here we describe the synthesis and multitarget biological profiling of a new class of compounds designed by molecular hybridization of an NMDA receptor antagonist fluorobenzohomoadamantanamine with the potent acetylcholinesterase (AChE) inhibitor 6-chlorotacrine, using two different linker lengths and linkage positions, to preserve or not the memantine-like polycyclic unsubstituted primary amine. The best hybrids exhibit greater potencies than parent compounds against AChE (IC50 0.33 nM in the best case, 44-fold increased potency over 6-chlorotacrine), butyrylcholinesterase (IC50 21 nM in the best case, 24-fold increased potency over 6-chlorotacrine), and NMDA receptors (IC50 0.89 μM in the best case, 2-fold increased potency over the parent benzohomoadamantanamine and memantine), which suggests an additive effect of both pharmacophoric moieties in the interaction with the primary targets. Moreover, most of these compounds have been predicted to be brain permeable. This set of biological properties makes them promising leads for further anti-Alzheimer drug development.
Collapse
Affiliation(s)
- F Javier Pérez-Areales
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain
| | - Andreea L Turcu
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain
| | - Marta Barniol-Xicota
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain
| | - Caterina Pont
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain
| | - Deborah Pivetta
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain
| | - Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Angela De Simone
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Corso D'Augusto 237, I-47921, Rimini, Italy
| | - Vincenza Andrisano
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, Corso D'Augusto 237, I-47921, Rimini, Italy
| | - Belén Pérez
- Department of Pharmacology, Therapeutics, and Toxicology, Autonomous University of Barcelona, E-08193, Bellaterra, Spain
| | - Raimon Sabate
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain
| | - Francesc X Sureda
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, C/St. Llorenç 21, E-43201, Reus, Spain
| | - Santiago Vázquez
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain.
| | - Diego Muñoz-Torrero
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028, Barcelona, Spain.
| |
Collapse
|
29
|
QuinoxalineTacrine QT78, a Cholinesterase Inhibitor as a Potential Ligand for Alzheimer's Disease Therapy. Molecules 2019; 24:molecules24081503. [PMID: 30999586 PMCID: PMC6514705 DOI: 10.3390/molecules24081503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/14/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022] Open
Abstract
We report the synthesis and relevant pharmacological properties of the quinoxalinetacrine (QT) hybrid QT78 in a project targeted to identify new non-hepatotoxic tacrine derivatives for Alzheimer’s disease therapy. We have found that QT78 is less toxic than tacrine at high concentrations (from 100 μM to 1 mM), less potent than tacrine as a ChE inhibitor, but shows selective BuChE inhibition (IC50 (hAChE) = 22.0 ± 1.3 μM; IC50 (hBuChE) = 6.79 ± 0.33 μM). Moreover, QT78 showed effective and strong neuroprotection against diverse toxic stimuli, such as rotenone plus oligomycin-A or okadaic acid, of biological significance for Alzheimer’s disease.
Collapse
|
30
|
AlFadly ED, Elzahhar PA, Tramarin A, Elkazaz S, Shaltout H, Abu-Serie MM, Janockova J, Soukup O, Ghareeb DA, El-Yazbi AF, Rafeh RW, Bakkar NMZ, Kobeissy F, Iriepa I, Moraleda I, Saudi MN, Bartolini M, Belal AS. Tackling neuroinflammation and cholinergic deficit in Alzheimer's disease: Multi-target inhibitors of cholinesterases, cyclooxygenase-2 and 15-lipoxygenase. Eur J Med Chem 2019; 167:161-186. [DOI: 10.1016/j.ejmech.2019.02.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 12/31/2022]
|
31
|
Design, synthesis, and evaluation of isoflavone analogs as multifunctional agents for the treatment of Alzheimer's disease. Eur J Med Chem 2019; 168:207-220. [DOI: 10.1016/j.ejmech.2019.02.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 12/11/2022]
|
32
|
Bartolini M, Marco‐Contelles J. Tacrines as Therapeutic Agents for Alzheimer's Disease. IV. The Tacripyrines and Related Annulated Tacrines. CHEM REC 2018; 19:927-937. [DOI: 10.1002/tcr.201800155] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Manuela Bartolini
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum University of Bologna Via Belmeloro 6 40126 Bologna Italy
| | - José Marco‐Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC) 3, Juan de la Cierva 28006 Madrid Spain
| |
Collapse
|
33
|
Prasher P, Sharma M. Medicinal chemistry of acridine and its analogues. MEDCHEMCOMM 2018; 9:1589-1618. [PMID: 30429967 PMCID: PMC6195008 DOI: 10.1039/c8md00384j] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 08/14/2018] [Indexed: 02/01/2023]
Abstract
'Acridine' along with its functional analogue 'Acridone' is the most privileged pharmacophore in medicinal chemistry with diverse applications ranging from DNA intercalators, endonuclease mimics, ratiometric selective ion sensors, and P-glycoprotein inhibitors in countering the multi-drug resistance, enzyme inhibitors, and reversals of neurodegenerative disorders. Their interaction with DNA and ability of selectively identifying numerous biologically useful ions has cemented exploitability of the acridone nucleus in modern day therapeutics. Additionally, most derivatives and salts of acridine are planar, crystalline, and stable displaying a strong fluorescence which, when coupled with their marked bio selectivity and low cytotoxicity, enables the studying and monitoring of several biochemical, metabolic, and pharmacological processes. In this review, a detailed picture covering the important therapeutic aspects of the acridone nucleus and its functional analogues is discussed.
Collapse
Affiliation(s)
- Parteek Prasher
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
- Department of Chemistry , University of Petroleum & Energy Studies , Dehradun 248007 , India . ;
| | - Mousmee Sharma
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
| |
Collapse
|
34
|
Girek M, Szymański P. Tacrine hybrids as multi-target-directed ligands in Alzheimer’s disease: influence of chemical structures on biological activities. CHEMICAL PAPERS 2018. [DOI: 10.1007/s11696-018-0590-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|